201
|
Liu S, Cao X, Wang D, Zhu H. Iron metabolism: State of the art in hypoxic cancer cell biology. Arch Biochem Biophys 2022; 723:109199. [DOI: 10.1016/j.abb.2022.109199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 02/08/2023]
|
202
|
Li Y, Li M, Liu L, Xue C, Fei Y, Wang X, Zhang Y, Cai K, Zhao Y, Luo Z. Cell-Specific Metabolic Reprogramming of Tumors for Bioactivatable Ferroptosis Therapy. ACS NANO 2022; 16:3965-3984. [PMID: 35200009 DOI: 10.1021/acsnano.1c09480] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Ferroptosis is a nonapoptotic iron-dependent cell death pathway with a significant clinical potential, but its translation is impeded by lack of tumor-specific ferroptosis regulators and aberrant tumor iron metabolism. Herein, we report a combinational strategy based on clinically tested constituents to selectively induce ferroptosis in metabolically reprogrammed tumor cells through cooperative GPX4-inhibition and ferritinophagy-enabled Fe2+ reinforcement. Azido groups were first introduced on tumor cells using biocompatible long-circulating self-assemblies based on polyethylene glycol-disulfide-N-azidoacetyl-d-mannosamine via metabolic glycoengineering. The azido-expressing tumor cells could specifically react with dibenzocyclooctyne-modified disulfide-bridged nanoassemblies via bioorthogonal click reactions, where the nanoassemblies were loaded with ferroptosis inducer RSL3 and ferritinophagy initiator dihydroartemisinin (DHA) and could release them in a bioresponsive manner. DHA-initiated ferritinophagy could degrade intracellular ferritin to liberate stored iron species and cooperate with the RSL3-mediated GPX4-inhibition for enhanced ferroptosis therapy. This tumor-specific ferroptosis induction strategy provides a generally applicable therapy with enhanced translatability, especially for tumors lacking targetable endogenous receptors.
Collapse
Affiliation(s)
- Yanan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Li Liu
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Chencheng Xue
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yang Fei
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Xuan Wang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yuchen Zhang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
203
|
Shi Q, Xia F, Wang Q, Liao F, Guo Q, Xu C, Wang J. Discovery and repurposing of artemisinin. Front Med 2022; 16:1-9. [PMID: 35290595 PMCID: PMC8922983 DOI: 10.1007/s11684-021-0898-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2022]
Abstract
Malaria is an ancient infectious disease that threatens millions of lives globally even today. The discovery of artemisinin, inspired by traditional Chinese medicine (TCM), has brought in a paradigm shift and been recognized as the “best hope for the treatment of malaria” by World Health Organization. With its high potency and low toxicity, the wide use of artemisinin effectively treats the otherwise drug-resistant parasites and helps many countries, including China, to eventually eradicate malaria. Here, we will first review the initial discovery of artemisinin, an extraordinary journey that was in stark contrast with many drugs in western medicine. We will then discuss how artemisinin and its derivatives could be repurposed to treat cancer, inflammation, immunoregulation-related diseases, and COVID-19. Finally, we will discuss the implications of the “artemisinin story” and how that can better guide the development of TCM today. We believe that artemisinin is just a starting point and TCM will play an even bigger role in healthcare in the 21st century.
Collapse
Affiliation(s)
- Qiaoli Shi
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Fei Xia
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qixin Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Fulong Liao
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qiuyan Guo
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Chengchao Xu
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China. .,Department of Geriatrics, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, 518020, China.
| | - Jigang Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China. .,Central People's Hospital of Zhanjiang, Zhanjiang, 524045, China. .,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China. .,Department of Geriatrics, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, 518020, China.
| |
Collapse
|
204
|
Yu T, Tan H, Liu C, Nie W, Wang Y, Zhou K, Shi H. Integratively Genomic Analysis Reveals the Prognostic and Immunological Characteristics of Pyroptosis and Ferroptosis in Pancreatic Cancer for Precision Immunotherapy. Front Cell Dev Biol 2022; 10:826879. [PMID: 35242763 PMCID: PMC8885993 DOI: 10.3389/fcell.2022.826879] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/05/2022] [Indexed: 02/05/2023] Open
Abstract
The non-apoptotic cell death processes including pyroptosis and ferroptosis have been implicated in the progression and therapeutic responses of pancreatic adenocarcinoma (PAAD). However, the extent to which pyroptosis and ferroptosis influence tumor biology remains ambiguous, especially in PAAD, which is characterized with “cold” immunity. Considering the heterogeneity among different patients, it was more practical to quantify distinct cell death profiles in an individual tumor sample. Herein, we developed a pyroptosis-ferroptosis (P-F) score for PAAD patients in The Cancer Genome Atlas (TCGA) database. A high P-F score was associated with active immune phenotype, decreased genomic alterations, and significantly longer survival. Good accuracy of the P-F score in predicting overall survival (OS) was further confirmed in the TCGA-PAAD, ICGC-PACA-CA, and E-MTAB-6134 cohorts. Besides, one immunotherapy cohort (IMvigor210 dataset) has verified that patients with high P-F scores exhibited significant advantages in therapeutic responses and clinical benefits. The sensitivity to chemotherapeutics was analyzed through the Genomics of Drug Sensitivity in Cancer (GDSC), and patients with low P-F score might be more sensitive to paclitaxel and 5-fluorouracil. Collectively, the P-F score based on the systematic evaluation of cell death profiles could serve as an effective biomarker in predicting the outcomes and responses of PAAD patients to treatments with chemotherapeutic agents or immunotherapies.
Collapse
Affiliation(s)
- Ting Yu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,Department of Pathology and Laboratory of Pathology, State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Huaicheng Tan
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chunhua Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Nie
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Wang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kexun Zhou
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
205
|
The Potential Mechanisms by which Artemisinin and Its Derivatives Induce Ferroptosis in the Treatment of Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1458143. [PMID: 35028002 PMCID: PMC8752222 DOI: 10.1155/2022/1458143] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/02/2021] [Accepted: 11/23/2021] [Indexed: 02/06/2023]
Abstract
Artemisinin (ART) is a bioactive molecule derived from the Chinese medicinal plant Artemisia annua (Asteraceae). ART and artemisinin derivatives (ARTs) have been effectively used for antimalaria treatment. The structure of ART is composed of a sesquiterpene lactone, including a peroxide internal bridge that is essential for its activity. In addition to their well-known antimalarial effects, ARTs have been shown recently to resist a wide range of tumors. The antineoplastic mechanisms of ART mainly include cell cycle inhibition, inhibition of tumor angiogenesis, DNA damage, and ferroptosis. In particular, ferroptosis is a novel nonapoptotic type of programmed cell death. However, the antitumor mechanisms of ARTs by regulating ferroptosis remain unclear. Through this review, we focus on the potential antitumor function of ARTs by acting on ferroptosis, including the regulation of iron metabolism, generation of reactive oxygen species (ROS), and activation of endoplasmic reticulum stress (ERS). This article systematically reviews the recent progress in ferroptosis research and provides a basis for ARTs as an anticancer drug in clinical practice.
Collapse
|
206
|
Superoxide Radicals in the Execution of Cell Death. Antioxidants (Basel) 2022; 11:antiox11030501. [PMID: 35326151 PMCID: PMC8944419 DOI: 10.3390/antiox11030501] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 12/24/2022] Open
Abstract
Superoxide is a primary oxygen radical that is produced when an oxygen molecule receives one electron. Superoxide dismutase (SOD) plays a primary role in the cellular defense against an oxidative insult by ROS. However, the resulting hydrogen peroxide is still reactive and, in the presence of free ferrous iron, may produce hydroxyl radicals and exacerbate diseases. Polyunsaturated fatty acids are the preferred target of hydroxyl radicals. Ferroptosis, a type of necrotic cell death induced by lipid peroxides in the presence of free iron, has attracted considerable interest because of its role in the pathogenesis of many diseases. Radical electrons, namely those released from mitochondrial electron transfer complexes, and those produced by enzymatic reactions, such as lipoxygenases, appear to cause lipid peroxidation. While GPX4 is the most potent anti-ferroptotic enzyme that is known to reduce lipid peroxides to alcohols, other antioxidative enzymes are also indirectly involved in protection against ferroptosis. Moreover, several low molecular weight compounds that include α-tocopherol, ascorbate, and nitric oxide also efficiently neutralize radical electrons, thereby suppressing ferroptosis. The removal of radical electrons in the early stages is of primary importance in protecting against ferroptosis and other diseases that are related to oxidative stress.
Collapse
|
207
|
Miao H, Ren Q, Li H, Zeng M, Chen D, Xu C, Chen Y, Wen Z. Comprehensive analysis of the autophagy-dependent ferroptosis-related gene FANCD2 in lung adenocarcinoma. BMC Cancer 2022; 22:225. [PMID: 35236309 PMCID: PMC8889748 DOI: 10.1186/s12885-022-09314-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 02/17/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The development of lung adenocarcinoma (LUAD) involves the interactions between cell proliferation and death. Autophagy-dependent ferroptosis, a distinctive cell death process, was implicated in a multitude of diseases, whereas no research revealing the relationship between autophagy-dependent ferroptosis and LUAD pathogenesis was reported. Thus, the primary objective was to explore the role and potential function of the autophagy-dependent ferroptosis-related genes in LUAD. METHODS Clinical information and transcriptome profiling of patients with LUAD were retrieved and downloaded from open-source databases. Autophagy-dependent ferroptosis-related genes were screened by published articles. The critical gene was identified as the intersection between the differentially expressed genes and prognosis-related genes. Patients were divided into high- and low-risk groups using the expression level of the critical gene. The validity of the key gene prognosis model was verified by survival analysis. The correlation between the clinical characteristics of LUAD and the expression level of the key gene was analyzed to explore the clinical significance and prognosis value. And the roles of the key gene in response to chemotherapy, immune microenvironment, and tumor mutation burden were predicted. The validation of key gene expression levels was further performed by quantitative real-time PCR and immunohistochemistry staining. RESULTS FANCD2, an essential autophagy-dependent ferroptosis-related gene by searching database, was confirmed as an independent prognostic factor for LUAD occurrence. The high expression level of FANCD2 was associated with an advantaged TNM stage, a less chemotherapy sensitivity, a low ImmuneScore, which indicated a deactivation status in an immune microenvironment, a high tumor mutation burden, and poor survival for LUAD patients. Pathway enrichment analysis showed that FANCD2 responded to oxidative stress and neutrophil-mediated immunity. Quantitative real-time PCR and immunohistochemistry staining showed that the expression level of FANCD2 is higher in LUAD patients than in normal tissue samples, which was in accordance with the database report. CONCLUSION FANCD2, an essential gene related to autophagy-dependent ferroptosis, could work as a biomarker, predicting the survival, chemotherapy sensitivity, tumor immunity, and mutation burden of LUAD. Researching autophagy-dependent ferroptosis and targeting the FANCD2 may offer a new perspective for treating and improving prognosis in LUAD.
Collapse
Affiliation(s)
- Huikai Miao
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfengdong, Guangzhou, 510060, People's Republic of China
| | - Qiannan Ren
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hongmu Li
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfengdong, Guangzhou, 510060, People's Republic of China
| | - Mingyue Zeng
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfengdong, Guangzhou, 510060, People's Republic of China
| | - Dongni Chen
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfengdong, Guangzhou, 510060, People's Republic of China
| | - Chunmei Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Youfang Chen
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfengdong, Guangzhou, 510060, People's Republic of China
| | - Zhesheng Wen
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfengdong, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
208
|
Shen M, Guo M, Li Y, Wang Y, Qiu Y, Shao J, Zhang F, Xu X, Yin G, Wang S, Chen A, Zhang Z, Zheng S. m 6A methylation is required for dihydroartemisinin to alleviate liver fibrosis by inducing ferroptosis in hepatic stellate cells. Free Radic Biol Med 2022; 182:246-259. [PMID: 35248719 DOI: 10.1016/j.freeradbiomed.2022.02.028] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 01/07/2023]
Abstract
Activation of hepatic stellate cells (HSCs) is a central event in the development of liver fibrosis, and the elimination of activated HSCs is considered to be an effective anti-fibrotic strategy. Here, we report that dihydroartemisinin (DHA) prevented the activation of HSCs via ferroptosis pathway. Importantly, DHA treatment increased the level of autophagy in HSCs. The inhibition of autophagy by 3-MA dramatically abolished the DHA-induced ferroptosis in HSCs. Mechanistically, the up-regulated m6A modification is essential for the activation of autophagy by DHA through the reduction of fat mass and obesity-associated gene (FTO). Down-regulation of m6A modification by FTO overexpression could impair autophagy and the classical ferroptotic events. Interestingly, the m6A modification of BECN1 mRNA was evidently up-regulated compared with other autophagy-related genes. More importantly, YTHDF1 was identified as a key m6A reader protein for BECN1 mRNA stability, and knockdown of YTHDF1 could prevent DHA-induced HSC ferroptosis. Noteworthy, YTH domain was essential for YTHDF1 to prolong the half-life of BECN1 mRNA in DHA-induced HSC ferroptosis. In mice, DHA treatment alleviated liver fibrosis by triggering HSC ferroptosis. HSC-specific inhibition of m6A modification and autophagy could impair DHA-induced HSC ferroptosis in murine liver fibrosis. Overall, these results provided novel implications to reveal the molecular mechanism of DHA-induced ferroptosis, by which pointed to m6A modification-dependent ferroptosis as a potential target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Min Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mei Guo
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yujia Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yingqian Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yangling Qiu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xuefen Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guoping Yin
- Department of Anesthesiology, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Shijun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250035, China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St Louis, MO, 63104, USA
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
209
|
Wang HT, Ju J, Wang SC, Zhang YH, Liu CY, Wang T, Yu X, Wang F, Cheng XR, Wang K, Chen ZY. Insights Into Ferroptosis, a Novel Target for the Therapy of Cancer. Front Oncol 2022; 12:812534. [PMID: 35280796 PMCID: PMC8914339 DOI: 10.3389/fonc.2022.812534] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/26/2022] [Indexed: 01/17/2023] Open
Abstract
Ferroptosis is a new form of programmed cell death (PCD) characterized by an excess iron accumulation and subsequent unbalanced redox states. Ferroptosis is different from the already reported PCD and has unique morphological features and biochemical processes. Ferroptosis was first elaborated by Brent R. Stockwell’s lab in 2012, in which small molecules erastin and RSL-3 induce PCD in Ras mutant cell lines. Ferroptosis involves various physiological processes and occurrence of disease and especially shows strong potential in cancer treatment. Development of small molecule compounds based on Stockwell’s research was found to kill cancer cells, and some FDA-approved drugs were discovered to result in ferroptosis of cancer cells. Radiotherapy and checkpoint therapy have been widely used as a treatment for many types of cancer. Recently, some papers have reported that chemotherapy, radiotherapy, and checkpoint therapy induce ferroptosis of cancer cells, which provides new strategies for cancer treatment. Nevertheless, the limitless proliferation of tumor cells and the lack of cell death mechanisms are important reasons for drug resistance for tumor therapy. Therefore, we reviewed the molecular mechanism of ferroptosis and sensitivity to ferroptosis of different cancer cells and tumor treatment strategy.
Collapse
Affiliation(s)
- Hong-Tao Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
- Science and Technology Department, Qingdao University, Qingdao, China
| | - Jie Ju
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Shao-Cong Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yu-Hui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Cui-Yun Liu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Tao Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xue Yu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Fei Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xue-Ru Cheng
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
- *Correspondence: Kun Wang, ; Zhao-Yang Chen,
| | - Zhao-Yang Chen
- Cardiology Department, Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, China
- *Correspondence: Kun Wang, ; Zhao-Yang Chen,
| |
Collapse
|
210
|
Lan H, Gao Y, Zhao Z, Mei Z, Wang F. Ferroptosis: Redox Imbalance and Hematological Tumorigenesis. Front Oncol 2022; 12:834681. [PMID: 35155264 PMCID: PMC8826956 DOI: 10.3389/fonc.2022.834681] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/03/2022] [Indexed: 01/19/2023] Open
Abstract
Ferroptosis is a novel characterized form of cell death featured with iron-dependent lipid peroxidation, which is distinct from any known programmed cell death in the biological processes and morphological characteristics. Recent evidence points out that ferroptosis is correlated with numerous metabolic pathways, including iron homeostasis, lipid metabolism, and redox homeostasis, associating with the occurrence and treatment of hematological malignancies, such as multiple myeloma, leukemia, and lymphoma. Nowadays, utilizing ferroptosis as the target to prevent and treat hematological malignancies has become an active and challenging topic of research, and the regulatory network and physiological function of ferroptosis also need to be further elucidated. This review will summarize the recent progress in the molecular regulation of ferroptosis and the physiological roles and therapeutic potential of ferroptosis as the target in hematological malignancies.
Collapse
Affiliation(s)
- Hongying Lan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yu Gao
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhengyang Zhao
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Ziqing Mei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Feng Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
211
|
Zhou X, Suo F, Haslinger K, Quax WJ. Artemisinin-Type Drugs in Tumor Cell Death: Mechanisms, Combination Treatment with Biologics and Nanoparticle Delivery. Pharmaceutics 2022; 14:395. [PMID: 35214127 PMCID: PMC8875250 DOI: 10.3390/pharmaceutics14020395] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Artemisinin, the most famous anti-malaria drug initially extracted from Artemisia annua L., also exhibits anti-tumor properties in vivo and in vitro. To improve its solubility and bioavailability, multiple derivatives have been synthesized. However, to reveal the anti-tumor mechanism and improve the efficacy of these artemisinin-type drugs, studies have been conducted in recent years. In this review, we first provide an overview of the effect of artemisinin-type drugs on the regulated cell death pathways, which may uncover novel therapeutic approaches. Then, to overcome the shortcomings of artemisinin-type drugs, we summarize the recent advances in two different therapeutic approaches, namely the combination therapy with biologics influencing regulated cell death, and the use of nanocarriers as drug delivery systems. For the former approach, we discuss the superiority of combination treatments compared to monotherapy in tumor cells based on their effects on regulated cell death. For the latter approach, we give a systematic overview of nanocarrier design principles used to deliver artemisinin-type drugs, including inorganic-based nanoparticles, liposomes, micelles, polymer-based nanoparticles, carbon-based nanoparticles, nanostructured lipid carriers and niosomes. Both approaches have yielded promising findings in vitro and in vivo, providing a strong scientific basis for further study and upcoming clinical trials.
Collapse
Affiliation(s)
| | | | - Kristina Haslinger
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (F.S.)
| | - Wim J. Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (F.S.)
| |
Collapse
|
212
|
Wu J, Ye J, Xie Q, Liu B, Liu M. Targeting Regulated Cell Death with Pharmacological Small Molecules: An Update on Autophagy-Dependent Cell Death, Ferroptosis, and Necroptosis in Cancer. J Med Chem 2022; 65:2989-3001. [PMID: 35130435 DOI: 10.1021/acs.jmedchem.1c01572] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulated cell death is a widely attractive subject among the topics of cancer therapy and has gained some advances for discovery of targeted anticancer drugs. In the past decade, nonapoptotic regulated cell death has been implicated in the development and therapeutic responses of a variety of human cancers. Hitherto, targeting autophagy-dependent cell death (ADCD), ferroptosis, and necroptosis with small molecules has been emerging as a hopeful strategy for the improvement of potential cancer therapy, which may have an advantage to bypass the apoptosis-resistance machinery. Thus, in this perspective, we concentrate on the key molecular insights into ADCD, ferroptosis, and necroptosis and summarize the corresponding small molecules in potential cancer therapy. Moreover, the relationships between the three subroutines and small molecules modulating the crosstalk are discussed. We believe that these inspiring findings would be advantageous to exploiting more potential targets and pharmacological small molecules in future cancer treatment.
Collapse
Affiliation(s)
- Junhao Wu
- State Key Laboratory of Biotherapy and Cancer Center and Department of Orthopedics, and Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Ye
- State Key Laboratory of Biotherapy and Cancer Center and Department of Orthopedics, and Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Xie
- State Key Laboratory of Biotherapy and Cancer Center and Department of Orthopedics, and Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center and Department of Orthopedics, and Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ming Liu
- State Key Laboratory of Biotherapy and Cancer Center and Department of Orthopedics, and Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
213
|
Zhi Y, Zhang S, Zi M, Wang Y, Liu Y, Zhang M, Shi L, Yan Q, Zeng Z, Xiong W, Zhi K, Gong Z. Potential applications of N 6 -methyladenosine modification in the prognosis and treatment of cancers via modulating apoptosis, autophagy, and ferroptosis. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1719. [PMID: 35114735 DOI: 10.1002/wrna.1719] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/13/2021] [Accepted: 01/11/2022] [Indexed: 12/30/2022]
Abstract
N6 -methyladenosine (m6 A) is one of the most abundant modifications determining the fate of RNA. Currently, m6 A modification is tightly connected with tumorigenesis and presents novel promise in clinical applications. Regulated cell death (RCD) is a programmed mechanism that plays a complicated role in malignant transition. Regarding the main forms of RCD, aberrant levels of m6 A modification have been detected during the progression of apoptosis, autophagy, ferroptosis, necroptosis, and pyroptosis in several diseases. However, few reviews have elucidated the correlation between m6 A-modified RCD and carcinogenesis. In this review, we summarize the regulators of m6 A methylation and their functions in carcinogenesis through an overview of m6 A-modified RCD. Additionally, we assume the potential role of m6 A modification regulators as novel biomarkers for chemotherapies and precision medicine. Furthermore, we review the controversies and conflicts in m6 A explorations and predict future orientations of m6 A-modified RCD for clinical applications. This article is categorized under: Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs.
Collapse
Affiliation(s)
- Yuan Zhi
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Moxin Zi
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yian Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yuhang Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Mi Zhang
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Lei Shi
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qijia Yan
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Keqian Zhi
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
214
|
Lin L, Zhang MX, Zhang L, Zhang D, Li C, Li YL. Autophagy, Pyroptosis, and Ferroptosis: New Regulatory Mechanisms for Atherosclerosis. Front Cell Dev Biol 2022; 9:809955. [PMID: 35096837 PMCID: PMC8793783 DOI: 10.3389/fcell.2021.809955] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder characterized by the gradual buildup of plaques within the vessel wall of middle-sized and large arteries. The occurrence and development of atherosclerosis and the rupture of plaques are related to the injury of vascular cells, including endothelial cells, smooth muscle cells, and macrophages. Autophagy is a subcellular process that plays an important role in the degradation of proteins and damaged organelles, and the autophagy disorder of vascular cells is closely related to atherosclerosis. Pyroptosis is a proinflammatory form of regulated cell death, while ferroptosis is a form of regulated nonapoptotic cell death involving overwhelming iron-dependent lipid peroxidation. Both of them exhibit distinct features from apoptosis, necrosis, and autophagy in morphology, biochemistry, and genetics. However, a growing body of evidence suggests that pyroptosis and ferroptosis interact with autophagy and participate in the development of cancers, degenerative brain diseases and cardiovascular diseases. This review updated the current understanding of autophagy, pyroptosis, and ferroptosis, finding potential links and their effects on atherogenesis and plaque stability, thus providing ways to develop new pharmacological strategies to address atherosclerosis and stabilize vulnerable, ruptured plaques.
Collapse
Affiliation(s)
- Lin Lin
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mu-Xin Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dan Zhang
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yun-Lun Li
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
215
|
Ge C, Zhang S, Mu H, Zheng S, Tan Z, Huang X, Xu C, Zou J, Zhu Y, Feng D, Aa J. Emerging Mechanisms and Disease Implications of Ferroptosis: Potential Applications of Natural Products. Front Cell Dev Biol 2022; 9:774957. [PMID: 35118067 PMCID: PMC8804219 DOI: 10.3389/fcell.2021.774957] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/09/2021] [Indexed: 01/09/2023] Open
Abstract
Ferroptosis, a newly discovered form of regulatory cell death (RCD), has been demonstrated to be distinct from other types of RCD, such as apoptosis, necroptosis, and autophagy. Ferroptosis is characterized by iron-dependent lipid peroxidation and oxidative perturbation, and is inhibited by iron chelators and lipophilic antioxidants. This process is regulated by specific pathways and is implicated in diverse biological contexts, mainly including iron homeostasis, lipid metabolism, and glutathione metabolism. A large body of evidence suggests that ferroptosis is interrelated with various physiological and pathological processes, including tumor progression (neuro)degenerative diseases, and hepatic and renal failure. There is an urgent need for the discovery of novel effective ferroptosis-modulating compounds, even though some experimental reagents and approved clinical drugs have been well documented to have anti- or pro-ferroptotic properties. This review outlines recent advances in molecular mechanisms of the ferroptotic death process and discusses its multiple roles in diverse pathophysiological contexts. Furthermore, we summarize chemical compounds and natural products, that act as inducers or inhibitors of ferroptosis in the prevention and treatment of various diseases. Herein, it is particularly highlighted that natural products show promising prospects in ferroptosis-associated (adjuvant) therapy with unique advantages of having multiple components, multiple biotargets and slight side effects.
Collapse
Affiliation(s)
- Chun Ge
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Sujie Zhang
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Huiwen Mu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shaojun Zheng
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhaoyi Tan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xintong Huang
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chen Xu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jianjun Zou
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yubing Zhu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- *Correspondence: Yubing Zhu, ; Dong Feng, ; Jiye Aa,
| | - Dong Feng
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Nanjing Southern Pharmaceutical Technology Co., Ltd., Nanjing, China
- *Correspondence: Yubing Zhu, ; Dong Feng, ; Jiye Aa,
| | - Jiye Aa
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- *Correspondence: Yubing Zhu, ; Dong Feng, ; Jiye Aa,
| |
Collapse
|
216
|
Ma Z, Woon CYN, Liu CG, Cheng JT, You M, Sethi G, Wong ALA, Ho PCL, Zhang D, Ong P, Wang L, Goh BC. Repurposing Artemisinin and its Derivatives as Anticancer Drugs: A Chance or Challenge? Front Pharmacol 2022; 12:828856. [PMID: 35035355 PMCID: PMC8758560 DOI: 10.3389/fphar.2021.828856] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022] Open
Abstract
Cancer has become a global health problem, accounting for one out of six deaths. Despite the recent advances in cancer therapy, there is still an ever-growing need for readily accessible new therapies. The process of drug discovery and development is arduous and takes many years, and while it is ongoing, the time for the current lead compounds to reach clinical trial phase is very long. Drug repurposing has recently gained significant attention as it expedites the process of discovering new entities for anticancer therapy. One such potential candidate is the antimalarial drug, artemisinin that has shown anticancer activities in vitro and in vivo. In this review, major molecular and cellular mechanisms underlying the anticancer effect of artemisinin and its derivatives are summarised. Furthermore, major mechanisms of action and some key signaling pathways of this group of compounds have been reviewed to explore potential targets that contribute to the proliferation and metastasis of tumor cells. Despite its established profile in malaria treatment, pharmacokinetic properties, anticancer potency, and current formulations that hinder the clinical translation of artemisinin as an anticancer agent, have been discussed. Finally, potential solutions or new strategies are identified to overcome the bottlenecks in repurposing artemisinin-type compounds as anticancer drugs.
Collapse
Affiliation(s)
- Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Clariis Yi-Ning Woon
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Chen-Guang Liu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Jun-Ting Cheng
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Mingliang You
- Hangzhou Cancer Institute, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, China.,Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Paul Chi-Lui Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Daping Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Peishi Ong
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Boon-Cher Goh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| |
Collapse
|
217
|
Song Y, Tian S, Zhang P, Zhang N, Shen Y, Deng J. Construction and Validation of a Novel Ferroptosis-Related Prognostic Model for Acute Myeloid Leukemia. Front Genet 2022; 12:708699. [PMID: 35111195 PMCID: PMC8803125 DOI: 10.3389/fgene.2021.708699] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 12/28/2021] [Indexed: 12/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal malignant proliferative blood disorder with a poor prognosis. Ferroptosis, a novel form of programmed cell death, holds great promise for oncology treatment, and has been demonstrated to interfere with the development of various diseases. A range of genes are involved in regulating ferroptosis and can serve as markers of it. Nevertheless, the prognostic significance of these genes in AML remains poorly understood. Transcriptomic and clinical data for AML patients were acquired from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). Univariate Cox analysis was performed to identify ferroptosis-related genes with prognostic value, and the least absolute shrinkage and selection operator (LASSO) algorithm and stepwise multivariate Cox regression analysis were utilized to optimize gene selection from the TCGA cohort (132 samples) for model construction. Tumor samples from the GEO database (136 samples and 104 samples) were used as validation groups to estimate the predictive performance of the risk model. Finally, an eight-gene prognostic signature (including CHAC1, CISD1, DPP4, GPX4, AIFM2, SQLE, PGD, and ACSF2) was identified for the prediction of survival probability and was used to stratify AML patients into high- and low-risk groups. Survival analysis illustrated significantly prolonged overall survival and lower mortality in the low-risk group. The area under the receiver operating characteristic curve demonstrated good results for the training set (1-year: 0.846, 2-years: 0.826, and 3-years: 0.837), which verified the accuracy of the model for predicting patient survival. Independent prognostic analysis indicated that the model could be used as a prognostic factor (p ≤ 0.001). Functional enrichment analyses revealed underlying mechanisms and notable differences in the immune status of the two risk groups. In brief, we conducted and validated a novel ferroptosis-related prognostic model for outcome prediction and risk stratification in AML, with great potential to guide individualized treatment strategies in the future.
Collapse
Affiliation(s)
- Ying Song
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shufang Tian
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Zhang
- Hematology Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nan Zhang
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Shen
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianchuan Deng
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Jianchuan Deng,
| |
Collapse
|
218
|
Yang F, Sun SY, Wang S, Guo JT, Liu X, Ge N, Wang GX. Molecular regulatory mechanism of ferroptosis and its role in gastrointestinal oncology: Progress and updates. World J Gastrointest Oncol 2022; 14:1-18. [PMID: 35116100 PMCID: PMC8790407 DOI: 10.4251/wjgo.v14.i1.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/04/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal (GI) tumors, including liver, pancreatic, gastric, and colorectal cancers, have a high incidence rate and low survival rate due to the lack of effective therapeutic methods and frequent relapses. Surgery and postoperative chemoradiotherapy have largely reduced the fatality rates for most GI tumors, but these therapeutic approaches result in poor prognoses due to severe adverse reactions and the development of drug resistance. Recent studies have shown that ferroptosis plays an important role in the onset and progression of GI tumors. Ferroptosis is a new non-apoptotic form of cell death, which is iron-dependent, non-apoptotic cell death characterized by the accumulation of lipid reactive oxygen species (ROS). The activation of ferroptosis can lead to tumor cell death. Thus, regulating ferroptosis in tumor cells may become a new therapeutic approach for tumors, making it become a research hotspot. Current studies suggest that ferroptosis is mainly triggered by the accumulation of lipid ROS. Furthermore, several studies have indicated that ferroptosis may be a new approach for the treatment of GI tumors. Here, we review current research progress on the mechanism of ferroptosis, current inducers and inhibitors of ferroptosis, and the role of ferroptosis in GI tumors to propose new methods for the treatment of such tumors.
Collapse
Affiliation(s)
- Fan Yang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Si-Yu Sun
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Sheng Wang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Jin-Tao Guo
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Xiang Liu
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Nan Ge
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Guo-Xin Wang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| |
Collapse
|
219
|
Nie Q, Hu Y, Yu X, Li X, Fang X. Induction and application of ferroptosis in cancer therapy. Cancer Cell Int 2022; 22:12. [PMID: 34996454 PMCID: PMC8742449 DOI: 10.1186/s12935-021-02366-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
At present, more than one cell death pathways have been found, one of which is ferroptosis. Ferroptosis was discovered in 2012 and described as an iron-dependent and lipid peroxidation-driven regulated cell death pathway. In the past few years, ferroptosis has been shown to induce tumor cell death, providing new ideas for tumor treatment. In this article, we summarize the latest advances in ferroptosis-induced tumor therapy at the intersection of tumor biology, molecular biology, redox biology, and materials chemistry. First, we state the characteristics of ferroptosis in cells, then introduce the key molecular mechanism of ferroptosis, and describes the relationship between ferroptosis and oxidative stress signaling pathways. Finally, we focused on several types of ferroptosis inducers discovered by scholars, and the application of ferroptosis in systemic chemotherapy, radiotherapy, immunotherapy and nanomedicine, in the hope that ferroptosis can exert its potential in the treatment of tumors.
Collapse
Affiliation(s)
- Qing Nie
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yue Hu
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xiao Yu
- First Affiliated Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xiao Li
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xuedong Fang
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China.
| |
Collapse
|
220
|
Wang X, Liu X, Wang H. Combination regimen of granulocyte colony-stimulating factor and recombinant human thrombopoietin improves the curative effect on elderly patients with leukemia through inducing pyroptosis and ferroptosis of leukemia cells. Cancer Gene Ther 2022; 29:1742-1750. [PMID: 35768562 PMCID: PMC9663303 DOI: 10.1038/s41417-022-00497-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/10/2022] [Accepted: 06/14/2022] [Indexed: 02/04/2023]
Abstract
Leukemia ranks as the one of most common causes of death from tumor. 51.4% of patients with leukemia are over 65 years old. However, the median overall survival (OS) of elderly leukemia patients is less than one year. It is urgent to explore more effective treatments for elderly patients with leukemia. Our recent prospective phase II single-arm study has revealed that combination regimen of granulocyte colony-stimulating factor (G-CSF) and recombinant human thrombopoietin (rhTPO) could improve the curative effect on elderly patients with leukemia, yet the precise mechanism remains unknown. This study demonstrated that combination of G-CSF and rhTPO showed greater effect on suppressing leukemia growth than G-CSF or rhTPO alone in vitro and in vivo. Mechanistically, G-CSF induced pyroptosis through ELANE in leukemia cells. Besides, rhTPO triggered ferroptosis by EP300 in leukemia cells. Moreover, rhTPO suppressed glutathione peroxidase 4 (GPX4) expression to induce ferroptosis through blocking the interaction between EP300 and GPX4 gene promoter via associating with EP300. In summary, this study illuminated that combination regimen of G-CSF and rhTPO improved the curative effect on elderly patients with leukemia through inducing pyroptosis and ferroptosis of leukemia cells. Therefore, our results provided a theoretical basis for combination regimen of G-CSF and rhTPO treating leukemia and potential therapeutic targets for leukemia.
Collapse
Affiliation(s)
- Xiaobin Wang
- grid.412449.e0000 0000 9678 1884Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110035 China
| | - Xiaoyu Liu
- grid.412449.e0000 0000 9678 1884Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110035 China
| | - Huihan Wang
- grid.412449.e0000 0000 9678 1884Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110035 China
| |
Collapse
|
221
|
Shao ZC, Zhu BH, Huang AF, Su MQ, An LJ, Wu ZP, Jiang YJ, Guo H, Han XQ, Liu CM. Docosahexaenoic Acid Reverses Epithelial-Mesenchymal Transition and Drug Resistance by Impairing the PI3K/AKT/ Nrf2/GPX4 Signalling Pathway in Docetaxel-Resistant PC3 Prostate Cancer Cells. Folia Biol (Praha) 2022; 68:59-71. [PMID: 36384263 DOI: 10.14712/fb2022068020059] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Drug resistance is a serious problem in cancer therapy. Growing evidence has shown that docosahexaenoic acid has anti-inflammatory and chemopreventive abilities. Studies have shown that autophagy inhibition and ferroptosis are promising therapeutic strategies for overcoming multidrug resistance. This study was aimed to examine whether docosahexaenoic acid (DHA) could reverse docetaxel resistance in prostate cancer cells. Cell survival was examined by MTT and colony formation. Protein expression was determined by Western blot. Reactive oxygen species (ROS) production was measured by flow cytometry. DHA displayed anti-cancer effects on proliferation, colony formation, migration, apoptosis, autophagy and epithelial mesenchymal transition. Glutathione-S-transferase π is an enzyme that plays an important role in drug resistance. DHA inhibited GSTπ protein expression and induced cytoprotective autophagy by regulating the PI3K/AKT signalling pathway in PC3R cells. DHA combined with PI3K inhibitor (LY294002) enhanced apoptosis by alleviating the expression of LC3B, (pro-) caspase- 3 and (uncleaved) PARP. DHA induced ferroptosis by attenuating the expression of glutathione peroxidase 4 (GPX4) and nuclear erythroid 2-related factor 2 (Nrf2). DHA-treated PC3R cells produced ROS. The ROS and cytotoxicity were reversed by treatment with ferrostatin-1. DHA combined with docetaxel inhibited EMT by regulating the expression of E-cadhein and N-cadherin. In summary, DHA reversed drug resistance and induced cytoprotective autophagy and ferroptosis by regulating the PI3K/AKT/Nrf2/GPX4 signalling pathway in PC3R cells. We propose that DHA could be developed as a chemosensitizer and that the PI3K/AKT /Nrf2/GPX4 signalling pathway might be a promising therapeutic target for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Z C Shao
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
- College of Chemistry and Bio-engineering, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| | - B H Zhu
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| | - A F Huang
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| | - M Q Su
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
- College of Chemistry and Bio-engineering, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| | - L J An
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
- College of Chemistry and Bio-engineering, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| | - Z P Wu
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
- School of Aesthetic Medicine, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| | - Y J Jiang
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| | - H Guo
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| | - X-Q Han
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| | - C-M Liu
- School of Medicine, Yichun University, Yuanzhou District, Yichun 336000, Jiangxi Province, China
| |
Collapse
|
222
|
Koike T, Takenaka M, Suzuki N, Ueda Y, Mori M, Hirayama T, Nagasawa H, Morishige KI. Intracellular ferritin heavy chain plays the key role in artesunate-induced ferroptosis in ovarian serous carcinoma cells. J Clin Biochem Nutr 2022; 71:34-40. [PMID: 35903602 PMCID: PMC9309081 DOI: 10.3164/jcbn.21-82] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/24/2021] [Indexed: 12/03/2022] Open
Abstract
Artesunate, an antimalarial drug, induces ferroptosis, but the mechanism is still unclear. In the present study, we investigated how Artesunate induces ferroptosis in ovarian serous carcinoma. Experiments were performed using the ovarian serous carcinoma cell lines CaOV3 and SKOV3ip1, and the sensitivity of CaOV3 to Artesunate was higher than that of SKOV3ip1. Ferroptosis inhibitors inhibited Artesunate-induced intracellular lipid peroxidation and cell death. However, unlike class 1 ferroptosis inducer erastin, Artesunate had no effect on intracellular glutathione-SH levels. We found that Artesunate-induced changes in lysosomal Fe2+ were parallel to the induction of ferroptosis. Therefore, ferritin, which oxidizes and binds intracellular Fe2+, may have an inhibitory effect on ferroptosis. Knockdown of nuclear coactivator 4, a key molecule of ferritinophagy (ferritin-specific autophagy), suppressed Artesunate-induced cell death. Knockdown of ferritin heavy chain by siRNA greatly enhanced the sensitivity to Artesunate, and overexpression of ferritin heavy chain greatly reduced the sensitivity of ovarian cancer cell lines to Artesunate. These results can explain the differential sensitivity of CaOV3 and SKOV3ip1 to Artesunate. In conclusion, enhancement of ferritinophagy is an important step involved in the mechanism of Artesunate-induced ferroptosis, and ferritin heavy chain levels may contribute to the regulation of sensitivity in Artesunate-induced ferroptosis in ovarian serous carcinoma cells.
Collapse
Affiliation(s)
- Tiger Koike
- Department of Obstetrics and Gynecology, Gifu University School of Medicine
| | - Motoki Takenaka
- Department of Obstetrics and Gynecology, Gifu University School of Medicine
| | - Noriko Suzuki
- Department of Obstetrics and Gynecology, Gifu University School of Medicine
| | - Yoko Ueda
- Department of Obstetrics and Gynecology, Gifu University School of Medicine
| | - Minako Mori
- Department of Obstetrics and Gynecology, Gifu University School of Medicine
| | - Tasuku Hirayama
- Laboratory of Pharmaceutical and Medical Chemistry, Gifu Pharmaceutical University
| | - Hideko Nagasawa
- Laboratory of Pharmaceutical and Medical Chemistry, Gifu Pharmaceutical University
| | | |
Collapse
|
223
|
FTH promotes the proliferation and renders the HCC cells specifically resist to ferroptosis by maintaining iron homeostasis. Cancer Cell Int 2021; 21:709. [PMID: 34965856 PMCID: PMC8717654 DOI: 10.1186/s12935-021-02420-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/19/2021] [Indexed: 02/06/2023] Open
Abstract
Background Ferroptosis is a newly identified type of programmed cell death, which preferentially targets iron-rich cancer cells such as hepatocellular carcinoma (HCC). Ferritin heavy chain (FTH) is a major iron storing nanocage to store redox-inactive iron, and harbors ferroxidase activity to prevent the iron-mediated production of ROS. Our previous studies have demonstrated that FTH acts as a protective role to increase the cellular resistance to ferroptosis. However, the specific role of FTH in the development of HCC and ferroptosis resistance remains unclear. Methods The indicated databases were used for bioinformatics analysis. The abilities of cell proliferation, migration were measured by cell proliferation assay, transwell assay and wound healing assay. The levels of reactive oxygen species (ROS), lipid peroxide, free iron, mitochondrial superoxide, mitochondrial morphology and mitochondrial membrane potential (MMP) were determined by DCF-DA, C11-BODIPY, mitoSOX, mitoTracker, JC-10 and TMRM staining, respectively. The mitochondrial oxygen consumption rate was monitored by the Seahorse XF24 Analyzer. Results The pan-cancer analysis was performed and showed that FTH expression is upregulated in multiple cancers, such as LIHC, CHOL, HNSC, compared to corresponding normal tissues. In addition, the level of serum ferritin is positively associated with the progression of hepatitis, cirrhosis liver and hepatocellular carcinoma. Further investigation shed light on the strong correlation between FTH expression and tumor grades, cancer stages and prognosis of HCC. Importantly, the proteins interaction network elucidated that FTH is involved in iron homeostasis maintenance and lysosomal-dependent degradation. Enforced expression of FTH accelerates proliferation, migration and endows HCC cells specifically resistant to ferroptosis, but does not protect against cell death caused by cytotoxic compounds like oxaliplatin, irinotecan, and adriamycin. Mechanically, FTH reconstituted cells exhibit diminished peroxides accumulation, reduce mitochondrial ROS level, attenuate the impaired mitochondrial respiratory and rescue the mitochondrial homeostasis. Notably, FTH expression boosts tumorigenic potential in vivo with increased PCNA staining and lesser lipid peroxides generation. Conclusion These results provide new insights that FTH acts as an oncogene in the carcinogenesis and progression of HCC, and is hopeful to be a potential target for therapeutic intervention through ferroptosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02420-x.
Collapse
|
224
|
Dihydroartemisinin Induces Ferroptosis in HCC by Promoting the Formation of PEBP1/15-LO. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3456725. [PMID: 34925691 PMCID: PMC8683180 DOI: 10.1155/2021/3456725] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/26/2021] [Accepted: 11/10/2021] [Indexed: 12/16/2022]
Abstract
Relevant researches have recognized the vital role of inducing ferroptosis in the treatment of tumor. The latest findings indicate that PEBP1/15-LO can play an essential role in the process of cell death. However, its role in regulating ferroptosis in hepatocellular carcinoma (simplified by HCC) remains unclear. The previous research of our team has proved that DHA can induce ferroptosis of hepatic stellate cells. In this study, we found that DHA could also induce ferroptosis in HCC cells. Interestingly, DHA induced ferroptosis by promoting the formation of PEBP1/15-LO and promoting cell membrane lipid peroxidation. In addition, we also found that DHA had no obvious regulatory effect on 15-LO, but it could promote PEBP1 protein expression. Importantly, we discovered the upregulation of PEBP1 induced by DHA was related to the inhibition of its ubiquitination degradation. In vivo experiments have also obtained consistent results that DHA can inhibit tumor growth and affect the expression of ferroptosis markers in tumor tissues, which would be partially offset by interference with PEBP1.
Collapse
|
225
|
Shi J, Wu P, Sheng L, Sun W, Zhang H. Ferroptosis-related gene signature predicts the prognosis of papillary thyroid carcinoma. Cancer Cell Int 2021; 21:669. [PMID: 34906147 PMCID: PMC8670268 DOI: 10.1186/s12935-021-02389-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer (TC), accounting for more than 80% of all cases. Ferroptosis is a novel iron-dependent and Reactive oxygen species (ROS) reliant type of cell death which is distinct from the apoptosis, necroptosis and pyroptosis. Considerable studies have demonstrated that ferroptosis is involved in the biological process of various cancers. However, the role of ferroptosis in PTC remains unclear. This study aims at exploring the expression of ferroptosis-related genes (FRG) and their prognostic values in PTC. METHODS A ferroptosis-related gene signature was constructed using lasso regression analysis through the PTC datasets of the Cancer Genome Atlas (TCGA). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to investigate the bioinformatics functions of significantly different genes (SDG) of ferroptosis. Additionally, the correlations of ferroptosis and immune cells were assessed through the single-sample gene set enrichment analysis (ssGSEA) and CIBERSORT database. Finally, SDG were test in clinical PTC specimens and normal thyroid tissues. RESULTS LASSO regression model was utilized to establish a novel FRG signature with 10 genes (ANGPTL7, CDKN2A, DPP4, DRD4, ISCU, PGD, SRXN1, TF, TFRC, TXNRD1) to predicts the prognosis of PTC, and the patients were separated into high-risk and low-risk groups by the risk score. The high-risk group had poorer survival than the low-risk group (p < 0.001). Receiver operating characteristic (ROC) curve analysis confirmed the signature's predictive capacity. Multivariate regression analysis identified the prognostic signature-based risk score was an independent prognostic indicator for PTC. The functional roles of the DEGs in the TGCA PTC cohort were explored using GO enrichment and KEGG pathway analyses. Immune related analysis demonstrated that the most types of immune cells and immunological function in the high-risk group were significant different with those in the low-risk group. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) verified the SDG have differences in expression between tumor tissue and normal thyroid tissue. In addition, cell experiments were conducted to observe the changes in cell morphology and expression of signature's genes with the influence of ferroptosis induced by sorafenib. CONCLUSIONS We identified differently expressed FRG that may involve in PTC. A ferroptosis-related gene signature has significant values in predicting the patients' prognoses and targeting ferroptosis may be an alternative for PTC's therapy.
Collapse
Affiliation(s)
- Jinyuan Shi
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Pu Wu
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Lei Sheng
- Department of Thyroid Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
226
|
Research progress on the occurrence and therapeutic mechanism of ferroptosis in NSCLC. Naunyn Schmiedebergs Arch Pharmacol 2021; 395:1-12. [PMID: 34779876 DOI: 10.1007/s00210-021-02178-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/06/2021] [Indexed: 10/19/2022]
Abstract
Ferroptosis refers to a novel way of cell death, inconsistent with the conventional concept of apoptosis and necrosis. It shows a close association with iron metabolism and oxidative damage, as marked by the significant increase of reactive oxygen species, the decreases of mitochondrial volume, and the thickening of membrane density. Recent studies confirmed that ferroptosis is closely associated with the occurrence, development, and therapy of the tumors. As impacted by the high levels of reactive oxygen species and lipid peroxides in lung cancer tissues, it is suggested that ferroptosis is more likely to occur in lung cancer tissues, which may act as a novel approach for non-small cell lung cancer (NSCLC) therapy. In the present study, the research achievements in recent years on the regulating mechanism of ferroptosis and its effect on the occurrence and the therapy of lung cancer are reviewed.
Collapse
|
227
|
Ferroptosis as a Major Factor and Therapeutic Target for Neuroinflammation in Parkinson's Disease. Biomedicines 2021; 9:biomedicines9111679. [PMID: 34829907 PMCID: PMC8615560 DOI: 10.3390/biomedicines9111679] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/16/2022] Open
Abstract
Mounting evidence suggests that ferroptosis is not just a consequence but also a fundamental contributor to the development and progression of Parkinson’s disease (PD). Ferroptosis is characterized as iron-dependent regulated cell death caused by excessive lipid peroxidation, leading to plasma membrane rupture, release of damage-associated molecular patterns, and neuroinflammation. Due to the crucial role of intracellular iron in mediating the production of reactive oxygen species and the formation of lipid peroxides, ferroptosis is intimately controlled by regulators involved in many aspects of iron metabolism, including iron uptake, storage and export, and by pathways constituting the antioxidant systems. Translational and transcriptional regulation of iron homeostasis and redox status provide an integrated network to determine the sensitivity of ferroptosis. We herein review recent advances related to ferroptosis, ranging from fundamental mechanistic discoveries and cutting-edge preclinical animal studies, to clinical trials in PD and the regulation of neuroinflammation via ferroptosis pathways. Elucidating the roles of ferroptosis in the survival of dopaminergic neurons and microglial activity can enhance our understanding of the pathogenesis of PD and provide opportunities for the development of novel prevention and treatment strategies.
Collapse
|
228
|
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: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [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
|
229
|
Liu L, Wang M, Gong N, Tian P, Deng H. Se improves GPX4 expression and SOD activity to alleviate heat-stress-induced ferroptosis-like death in goat mammary epithelial cells. Anim Cells Syst (Seoul) 2021; 25:283-295. [PMID: 34745435 PMCID: PMC8567913 DOI: 10.1080/19768354.2021.1988704] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Selenium (Se) is a vital element of life, which has an important impact on the growth, development, production performance and stress-tolerance of animals. However, it is not entirely clear that how exactly Se works during these processes. Herein, we investigate the role of Se in regulating the functions of goat mammary epithelial cells (GMECs) under heat-stress condition. We found that heat stress caused ferroptosis-like death in GMECs, manifested by a robust increase in iron ion concentration, reactive oxygen species (ROS) and cell death ratio, and a decrease in the activity of superoxide dismutase (SOD) and expression level of glutathione peroxidases 4 (GPX4). Se incubation had no obvious effect on GMEC viability, but alleviated heat-stress-induced ferroptosis-like cell death and improved GPX4 expression and SOD activity in a dose-dependent manner. Also, we found that overexpression of GPX4 could improve the activity of SOD. And Se incubation inhibited activation of mTOR signaling in heat-stress-induced GMECs, which could be eliminated by the mTOR activator MHY1485, and treatment with mTOR inhibitor AY-22989 had the same effect as Se. In conclusion, Se improves GPX4 expression and SOD activity and inhibits the activation of mTOR to alleviate heat-stress-induced ferroptosis-like death in GMECs, which may be a protective agent for heat stress in goats.
Collapse
Affiliation(s)
- Lu Liu
- College of Chemistry & Pharmacy, Northwest Agricultural & Forestry University, Yangling, People's Republic of China
| | - Manjiang Wang
- Fuping County Animal Epidemic Prevention Control Center, Xianyang, People's Republic of China
| | - Ning Gong
- College of Chemistry & Pharmacy, Northwest Agricultural & Forestry University, Yangling, People's Republic of China
| | - Peng Tian
- College of Chemistry & Pharmacy, Northwest Agricultural & Forestry University, Yangling, People's Republic of China
| | - Hongxia Deng
- College of Chemistry & Pharmacy, Northwest Agricultural & Forestry University, Yangling, People's Republic of China
| |
Collapse
|
230
|
Wu S, Zhu C, Tang D, Dou QP, Shen J, Chen X. The role of ferroptosis in lung cancer. Biomark Res 2021; 9:82. [PMID: 34742351 PMCID: PMC8572460 DOI: 10.1186/s40364-021-00338-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/20/2021] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is one of the most common cancers in the world. Although medical treatment has made impressive progress in recent years, it is still one of the leading causes of cancer-related deaths in men and women. Ferroptosis is a type of non-apoptotic cell death modality, usually characterized by iron-dependent lipid peroxidation, rather than caspase-induced protein cleavage. Excessive or lack of ferroptosis is associated with a variety of diseases, including cancer and ischaemia-reperfusion injury. Recent preclinical evidence suggests that targeting ferroptotic pathway is a potential strategy for the treatment of lung cancer. In this review, we summarize the core mechanism and regulatory network of ferroptosis in lung cancer cells, and highlight ferroptosis induction-related tumor therapies. The reviewed information may provide new insights for targeted lung cancer therapy.
Collapse
Affiliation(s)
- Sikai Wu
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, China
| | - Chengchu Zhu
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Q Ping Dou
- Department of Oncology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Departments of Pharmacology & Pathology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Jianfei Shen
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China.
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, China.
| | - Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
231
|
Wei J, Nai GY, Dai Y, Huang XJ, Xiong MY, Yao XY, Huang ZN, Li SN, Zhou WJ, Huang Y, Cheng P, Deng DH. Dipetidyl peptidase-4 and transferrin receptor serve as prognostic biomarkers for acute myeloid leukemia. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1381. [PMID: 34733933 PMCID: PMC8506534 DOI: 10.21037/atm-21-3368] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/29/2021] [Indexed: 01/22/2023]
Abstract
Background Acute myeloid leukemia (AML) is the most common hematological malignancy in adult patients. Ferroptosis-related signatures have been shown to act as regulators of the progression of multiple cancer types, but the role of ferroptosis in AML remains to be elucidated. We performed the present study to preliminarily investigate the roles of ferroptosis-related genes (FRGs) in AML. Methods The transcriptome data of AML patients was downloaded from The Cancer Genome Atlas (TCGA) and the transcriptome data of normal samples was obtained from the Genotype-Tissue Expression (GTEx) database. FRGs were selected via public articles. Expression levels of FRGs between AML and normal samples were analyzed. The prognostic model based on FRGs was constructed via lasso regression. The expression levels and prognostic role of FRGs were identified from the risk model. We also performed validation experiments to verify the expression levels of the final selected genes via immunohistochemistry, polymerase chain reaction (PCR), and RNA-seq. Finally, we explored the associations between immune infiltration, drug sensitivity, and the selected FRGs. Results The transcriptome data of 151 AML samples were retrieved from TCGA and 70 bone marrow normal samples were retrieved from the GTEx database. Additionally, 23 FRGs were collected from the published articles. There were 22 differentially expressed FRGs, and among them, dipetidyl peptidase-4 (DPP4) (P= 0.011, HR =1.504), GPX4 (P=0.055, HR =1.569), LPCAT3 (P<0.001, HR =2.243), SLC7A11 (P=0.012, HR =2.243), and transferrin receptor (TFRC) (P=0.029, 0.774) had a significant influence on the prognosis of AML patients via lasso regression. The area under the curve (AUC) values of the 1-, 3-, and 5-year receiver operating characteristic (ROC) curves of the FRG signatures indicated that this model is novel and effective method for predicting the prognosis of AML patients. DPP4 (P<0.001) was overexpressed while LPCAT3 (P<0.001), TFRC (P<0.001), GPX4 (P<0.001), and SLC7A11 (P<0.001) were downregulated, further validation experiment results indicated that DPP4 was significantly downregulated but TFRC was upregulated in AML samples. Dysregulation of DPP4 and TFRC influence numbers of chemotherapy regimens sensitivity. Conclusions DPP4 and TFRC act as biomarkers for predicting and diagnosing AML, and their expression levels also have significant correlations with drug resistance in AML.
Collapse
Affiliation(s)
- Jie Wei
- Department of Hematology, Baise People's Hospital, Baise, China
| | - Guan Ye Nai
- Department of hematology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yi Dai
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xun Jun Huang
- Department of Hematology, Baise People's Hospital, Baise, China
| | - Ming Yue Xiong
- Department of Hematology, Baise People's Hospital, Baise, China
| | - Xiang You Yao
- Department of Hematology, Baise People's Hospital, Baise, China
| | - Zhi Ning Huang
- Department of Hematology, Baise People's Hospital, Baise, China
| | - Si Nian Li
- Department of Hematology, Baise People's Hospital, Baise, China
| | - Wei Jie Zhou
- Department of Clinical Laboratory, Baise People's Hospital, Baise, China
| | - Yan Huang
- Department of Hematology, Baise People's Hospital, Baise, China
| | - Peng Cheng
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dong Hong Deng
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
232
|
Cosialls E, El Hage R, Dos Santos L, Gong C, Mehrpour M, Hamaï A. Ferroptosis: Cancer Stem Cells Rely on Iron until "to Die for" It. Cells 2021; 10:cells10112981. [PMID: 34831207 PMCID: PMC8616391 DOI: 10.3390/cells10112981] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer stem cells (CSCs) are a distinct subpopulation of tumor cells with stem cell-like features. Able to initiate and sustain tumor growth and mostly resistant to anti-cancer therapies, they are thought responsible for tumor recurrence and metastasis. Recent accumulated evidence supports that iron metabolism with the recent discovery of ferroptosis constitutes a promising new lead in the field of anti-CSC therapeutic strategies. Indeed, iron uptake, efflux, storage and regulation pathways are all over-engaged in the tumor microenvironment suggesting that the reprogramming of iron metabolism is a crucial occurrence in tumor cell survival. In particular, recent studies have highlighted the importance of iron metabolism in the maintenance of CSCs. Furthermore, the high concentration of iron found in CSCs, as compared to non-CSCs, underlines their iron addiction. In line with this, if iron is an essential macronutrient that is nevertheless highly reactive, it represents their Achilles’ heel by inducing ferroptosis cell death and therefore providing opportunities to target CSCs. In this review, we first summarize our current understanding of iron metabolism and its regulation in CSCs. Then, we provide an overview of the current knowledge of ferroptosis and discuss the role of autophagy in the (regulation of) ferroptotic pathways. Finally, we discuss the potential therapeutic strategies that could be used for inducing ferroptosis in CSCs to treat cancer.
Collapse
Affiliation(s)
- Emma Cosialls
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
| | - Rima El Hage
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
| | - Leïla Dos Santos
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
| | - Chang Gong
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Guangzhou 510120, China;
| | - Maryam Mehrpour
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
- Correspondence: (M.M.); (A.H.)
| | - Ahmed Hamaï
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
- Correspondence: (M.M.); (A.H.)
| |
Collapse
|
233
|
Yang D, Wang J, Li C, Shi L, Zhang M. Ferroptosis-related gene model to predict overall survival of papillary thyroid carcinoma. Am J Otolaryngol 2021; 42:103163. [PMID: 34339960 DOI: 10.1016/j.amjoto.2021.103163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/17/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Ferroptosis is a form of programmed cell death that is closely associated with the development of various tumors. However, the correlation between ferroptosis and papillary thyroid carcinoma (PTC) is unclear. This study was performed to investigate the expression and prognostic value of ferroptosis-related genes (FRG) in PTC. METHODS mRNA expression profiles and corresponding clinical data of patients with PTC were analyzed to identify factors affecting prognosis. Independent risk factors were used to establish a predictive receiver operating characteristic model. Single-sample gene set enrichment analysis (ssGSEA) was used to evaluate the correlation between ferroptosis and immune cells. RESULTS Most genes related to FRG (78.8%) were differentially expressed between the tumor and adjacent normal tissues. In univariate Cox regression analysis, 12 differentially expressed genes were associated with prognostic survival. We constructed a prognostic model of eight FRG, including DPP4, GPX4, GSS, ISCU, MIOX, PGD, TF, and TFRC, and divided patients into two groups: high and low risk. The high-risk group exhibited a significantly reduced overall survival rate. In multivariate Cox regression analysis, the risk score was used as an independent prognostic factor. ssGSEA showed that immune cell types and their expression in the high- and low-risk groups were significant. CONCLUSION This study constructed a prognostic model of ferroptosis-related genes and determined its usefulness as an independent prognostic factor, providing a reference for the treatment and prognosis of patients with PTC.
Collapse
Affiliation(s)
- Dianmei Yang
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Junwen Wang
- Department of Neurology, The Second People's Hospital of Guiyang, Guiyang 550081, Guizhou, China
| | - Chunyu Li
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Lixin Shi
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Miao Zhang
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China.
| |
Collapse
|
234
|
Park JM, Mau CZ, Chen YC, Su YH, Chen HA, Huang SY, Chang JS, Chiu CF. A case-control study in Taiwanese cohort and meta-analysis of serum ferritin in pancreatic cancer. Sci Rep 2021; 11:21242. [PMID: 34711879 PMCID: PMC8553768 DOI: 10.1038/s41598-021-00650-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most lethal diseases which lack an early diagnostic marker. We investigated whether serum ferritin (SF) reflects risk for pancreatic cancer and potential genes that may contribute ferritin and pancreatic cancer risks. We performed a meta-analysis of relevant studies on SF and pancreatic cancer risk by searching articles in PUBMED and EMBASE published up to 1 March 2020. We also collected serum samples from Taipei Medical University Joint Biobank and compared SF levels in 34 healthy controls and 34 pancreatic cancer patients. An Oncomine database was applied as a platform to explore a series of genes that exhibited strong associations between ferritin and pancreatic cancer. Herein, we show that high levels of SF can indicate risk of pancreatic cancer, suggesting SF as the new tumor marker that may be used to help pancreatic cancer diagnosis. We also found that expressions of iron homeostasis genes (MYC, FXN) and ferroptosis genes (ALOX15, CBS, FDFT1, LPCAT3, RPL8, TP53, TTC35) are significantly altered with pancreatic tumor grades, which may contribute to differential expression of ferritin related to pancreatic cancer prognosis.
Collapse
Affiliation(s)
- Ji Min Park
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan.,Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chen-Zou Mau
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yang-Ching Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan.,Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yen-Hao Su
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11301, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.,Department of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11301, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hsin-An Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11301, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.,Department of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11301, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan.,Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan.,Nutrition Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Jung-Su Chang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan. .,Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan. .,Nutrition Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
| | - Ching-Feng Chiu
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan. .,Nutrition Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan. .,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
235
|
Zhao Y, Huang Z, Peng H. Molecular Mechanisms of Ferroptosis and Its Roles in Hematologic Malignancies. Front Oncol 2021; 11:743006. [PMID: 34778060 PMCID: PMC8582018 DOI: 10.3389/fonc.2021.743006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
Cell death is essential for the normal metabolism of human organisms. Ferroptosis is a unique regulated cell death (RCD) mode characterized by excess accumulation of iron-dependent lipid peroxide and reactive oxygen species (ROS) compared with other well-known programmed cell death modes. It has been currently recognized that ferroptosis plays a rather important role in the occurrence, development, and treatment of traumatic brain injury, stroke, acute kidney injury, liver damage, ischemia-reperfusion injury, tumor, etc. Of note, ferroptosis may be explained by the expression of various molecules and signaling components, among which iron, lipid, and amino acid metabolism are the key regulatory mechanisms of ferroptosis. Meanwhile, tumor cells of hematological malignancies, such as leukemia, lymphoma, and multiple myeloma (MM), are identified to be sensitive to ferroptosis. Targeting potential regulatory factors in the ferroptosis pathway may promote or inhibit the disease progression of these malignancies. In this review, a systematic summary was conducted on the key molecular mechanisms of ferroptosis and the current potential relationships of ferroptosis with leukemia, lymphoma, and MM. It is expected to provide novel potential therapeutic approaches and targets for hematological malignancies.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Hematology, The Second Xiangya Hospital, Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
- Institute of Hematology, Central South University, Changsha, China
| | - Zineng Huang
- Department of Hematology, The Second Xiangya Hospital, Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
- Institute of Hematology, Central South University, Changsha, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
- Institute of Hematology, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, China
| |
Collapse
|
236
|
Yu R, Jin G, Fujimoto M. Dihydroartemisinin: A Potential Drug for the Treatment of Malignancies and Inflammatory Diseases. Front Oncol 2021; 11:722331. [PMID: 34692496 PMCID: PMC8529146 DOI: 10.3389/fonc.2021.722331] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022] Open
Abstract
Dihydroartemisinin (DHA) has been globally recognized for its efficacy and safety in the clinical treatment of malaria for decades. Recently, it has been found that DHA inhibits malignant tumor growth and regulates immune system function in addition to anti-malaria. In parasites and tumors, DHA causes severe oxidative stress by inducing excessive reactive oxygen species production. DHA also kills tumor cells by inducing programmed cell death, blocking cell cycle and enhancing anti-tumor immunity. In addition, DHA inhibits inflammation by reducing the inflammatory cells infiltration and suppressing the production of pro-inflammatory cytokines. Further, genomics, proteomics, metabolomics and network pharmacology of DHA therapy provide the basis for elucidating the pharmacological effects of DHA. This review provides a summary of the recent research progress of DHA in anti-tumor, inhibition of inflammatory diseases and the relevant pharmacological mechanisms. With further research of DHA, it is likely that DHA will become an alternative therapy in the clinical treatment of malignant tumors and inflammatory diseases.
Collapse
Affiliation(s)
- Ran Yu
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Guihua Jin
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Manabu Fujimoto
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Laboratory of Cutaneous Immunology, Osaka University Immunology Frontier Research Center, Osaka, Japan
| |
Collapse
|
237
|
Xuan Z, Zhang Y, Pan Z, Zheng X, Huang P. Natural medicinal ingredients induce tumor ferroptosis and related mechanisms. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:601-606. [PMID: 34986533 DOI: 10.3724/zdxbyxb-2021-0198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ferroptosis is an iron-dependent programmed cell death characterized by reactive oxygen species-induced lipid peroxide accumulation, which is different from cell apoptosis, pyroptosis, necrosis or autophagy. Ferroptosis plays an important role in the regulation of tumorigenesis and tumor development. Recent studies have shown that natural medicinal ingredients can induce ferroptosis in tumor cells through glutathione (GSH)/glutathione peroxidase 4 (GPx4) pathway, iron metabolism, lipid metabolism or other mechanisms. It has been reported that more than 30 natural medicinal ingredients can induce ferroptosis in tumor cells with multiple pathways and multiple targets. This article reviews the current research progress on the antitumor effects of natural medicinal ingredients through inducing cell ferroptosis.
Collapse
Affiliation(s)
- Zixue Xuan
- Department of Pharmacy, Clinical Pharmacy Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Yiwen Zhang
- Department of Pharmacy, Clinical Pharmacy Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Zongfu Pan
- Department of Pharmacy, Clinical Pharmacy Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Xiaowei Zheng
- Department of Pharmacy, Clinical Pharmacy Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Ping Huang
- Department of Pharmacy, Clinical Pharmacy Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| |
Collapse
|
238
|
Yang C, Li Y, Hu W, Wang X, Hu J, Yuan C, Zhou C, Wang H, Du J, Wang Y, Tong X. TEOA Promotes Autophagic Cell Death via ROS-Mediated Inhibition of mTOR/p70S6k Signaling Pathway in Pancreatic Cancer Cells. Front Cell Dev Biol 2021; 9:734818. [PMID: 34692691 PMCID: PMC8526869 DOI: 10.3389/fcell.2021.734818] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer is a common malignant tumor with high mortality, and novel therapeutic options have focused on ameliorating its poor prognosis. TEOA, a traditional Chinese herbal medicine, exhibits anti-inflammatory and anti-cancer activities. Our recent study has shown that TEOA inhibits proliferation and induces DNA damage in diffuse large B-cell lymphoma cells by activating the ROS-mediated p38 MAPK pathway. However, its effects on pancreatic cancer cells remain unknown. In the present study, we evaluated the effects of TEOA on the proliferation, migration of pancreatic cancer cells and explored the possible underlying mechanism of action. We found that TEOA significantly inhibited the proliferation and migration of pancreatic cancer cells in a time- and dose-dependent manner. Mechanistically, TEOA significantly induced mitochondrial dysfunction in PANC1 and SW1990 cells, as evidenced by the collapse of the mitochondrial membrane potential, exhausted ATP level, and excessive accumulation of intracellular ROS. Notably, our further experiments showed that TEOA induced autophagic cell death in pancreatic ductal adenocarcinoma cells by inactivating the ROS-dependent mTOR/p70S6k signaling pathway. More importantly, both pharmacological or genetic blocking of the autophagic flux signal could partly restore the cytotoxicity of TEOA, whereas activation of autophagy by rapamycin or EBSS induced starvation facilitated the cytotoxicity of TEOA. Concomitantly, N-acetylcysteine, a ROS scavenger, abolished the inhibition of the mTOR signaling pathway, thus preventing autophagy and restoring cell viability. Taken together, our results reveal that TEOA can lead to ROS-dependent autophagic cell death of pancreatic cancer cells by inducing mitochondrial dysfunction, which might be a promising therapeutic agent for pancreatic cancer.
Collapse
Affiliation(s)
- Chen Yang
- Department of Ultrasound, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wanye Hu
- Graduate School, Bengbu Medical College, Bengbu, China
| | - Xu Wang
- Laboratory Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Jiayu Hu
- Graduate School, Bengbu Medical College, Bengbu, China
| | - Chen Yuan
- Graduate School, Bengbu Medical College, Bengbu, China
| | - Chaoting Zhou
- Department of Ultrasound, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Hairui Wang
- Laboratory Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Jing Du
- Laboratory Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Ying Wang
- Graduate School, Bengbu Medical College, Bengbu, China
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
- Clinical Pharmacy Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xiangmin Tong
- Department of Ultrasound, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate School, Bengbu Medical College, Bengbu, China
- Laboratory Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
- Clinical Pharmacy Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
239
|
Liu L, Li L, Li M, Luo Z. Autophagy-Dependent Ferroptosis as a Therapeutic Target in Cancer. ChemMedChem 2021; 16:2942-2950. [PMID: 34110079 DOI: 10.1002/cmdc.202100334] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Indexed: 12/26/2022]
Abstract
Ferroptosis is an iron-dependent form of cell death associated with the accumulation of labile iron and cytotoxic lipid peroxides. Increasing evidence reveals that ferroptosis is not a self-standing phenomenon and has close connections with other cellular events. Remarkably, recent insights show that ferroptosis is dependent on autophagy, which is a lysosomal degradation pathway responsible for the recycling of damaged cellular components under survival stress. Autophagy is capable of contributing to ferroptosis through degradation of the ferritin, an iron-storage protein, accompanied with the accumulation of iron levels and lipid ROS. The interplay between autophagy and ferroptosis also reveals emerging opportunities for novel tumor therapies, which has inspired the development of many treatment strategies capable of inducing ferroptosis in tumor cells via autophagic pathways based on molecular and nanoparticulate agents. In this review, we summarize the specific molecular and regulatory networks of autophagy-dependent ferroptosis and highlight their pathophysiological impact on various aspects of tumor cells. A perspective was also provided regarding the preliminary therapeutic exploitation of ferroptosis/autophagy crosstalk for tumor treatment.
Collapse
Affiliation(s)
- Li Liu
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Liqi Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
240
|
Siddiqui AJ, Khan MF, Hamadou WS, Goyal M, Jahan S, Jamal A, Ashraf SA, Sharma P, Sachidanandan M, Badraoui R, Chaubey KK, Snoussi M, Adnan M. Molecular Docking and Dynamics Simulation Revealed Ivermectin as Potential Drug against Schistosoma-Associated Bladder Cancer Targeting Protein Signaling: Computational Drug Repositioning Approach. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:1058. [PMID: 34684095 PMCID: PMC8539496 DOI: 10.3390/medicina57101058] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/26/2021] [Accepted: 09/29/2021] [Indexed: 12/29/2022]
Abstract
Urogenital schistosomiasis is caused by Schistosoma haematobium (S. haematobium) infection, which has been linked to the development of bladder cancer. In this study, three repurposing drugs, ivermectin, arteether and praziquantel, were screened to find the potent drug-repurposing candidate against the Schistosoma-associated bladder cancer (SABC) in humans by using computational methods. The biology of most glutathione S-transferases (GSTs) proteins and vascular endothelial growth factor (VEGF) is complex and multifaceted, according to recent evidence, and these proteins actively participate in many tumorigenic processes such as cell proliferation, cell survival and drug resistance. The VEGF and GSTs are now widely acknowledged as an important target for antitumor therapy. Thus, in this present study, ivermectin displayed promising inhibition of bladder cancer cells via targeting VEGF and GSTs signaling. Moreover, molecular docking and molecular dynamics (MD) simulation analysis revealed that ivermectin efficiently targeted the binding pockets of VEGF receptor proteins and possessed stable dynamics behavior at binding sites. Therefore, we proposed here that these compounds must be tested experimentally against VEGF and GST signaling in order to control SABC. Our study lies within the idea of discovering repurposing drugs as inhibitors against the different types of human cancers by targeting essential pathways in order to accelerate the drug development cycle.
Collapse
Affiliation(s)
- Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail 2440, Saudi Arabia; (W.S.H.); (A.J.); (R.B.); (M.S.); (M.A.)
| | - Mohammad Faheem Khan
- Department of Biotechnology, Era’s Lucknow Medical College, Era University, Lucknow 226003, India;
| | - Walid Sabri Hamadou
- Department of Biology, College of Science, University of Hail, Hail 2440, Saudi Arabia; (W.S.H.); (A.J.); (R.B.); (M.S.); (M.A.)
| | - Manish Goyal
- Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; (M.G.); (P.S.)
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia;
| | - Arshad Jamal
- Department of Biology, College of Science, University of Hail, Hail 2440, Saudi Arabia; (W.S.H.); (A.J.); (R.B.); (M.S.); (M.A.)
| | - Syed Amir Ashraf
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Hail, Hail 2440, Saudi Arabia;
| | - Pankaj Sharma
- Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; (M.G.); (P.S.)
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Manojkumar Sachidanandan
- Department of Oral Radiology, College of Dentistry, University of Hail, Hail 2440, Saudi Arabia;
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Hail, Hail 2440, Saudi Arabia; (W.S.H.); (A.J.); (R.B.); (M.S.); (M.A.)
- Section of Histology-Cytology, Medicine Faculty of Tunis, University of Tunis El Manar, Tunis 1017, Tunisia
| | - Kundan Kumar Chaubey
- Department of Biotechnology, Academic Block VI, GLA University, Mathura 281406, India;
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Hail, Hail 2440, Saudi Arabia; (W.S.H.); (A.J.); (R.B.); (M.S.); (M.A.)
- Laboratory of Genetics, Biodiversity and Valorization of Bio-Resources (LR11ES41), University of Monastir, Higher Institute of Biotechnology of Monastir, Avenue Tahar Haddas BP74, Monastir 5000, Tunisia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail 2440, Saudi Arabia; (W.S.H.); (A.J.); (R.B.); (M.S.); (M.A.)
| |
Collapse
|
241
|
Chen X, Kang R, Kroemer G, Tang D. Organelle-specific regulation of ferroptosis. Cell Death Differ 2021; 28:2843-2856. [PMID: 34465893 PMCID: PMC8481335 DOI: 10.1038/s41418-021-00859-z] [Citation(s) in RCA: 215] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023] Open
Abstract
Ferroptosis, a cell death modality characterized by iron-dependent lipid peroxidation, is involved in the development of multiple pathological conditions, including ischemic tissue damage, infection, neurodegeneration, and cancer. The cellular machinery responsible for the execution of ferroptosis integrates multiple pro-survival or pro-death signals from subcellular organelles and then 'decides' whether to engage the lethal process or not. Here, we outline the evidence implicating different organelles (including mitochondria, lysosomes, endoplasmic reticulum, lipid droplets, peroxisomes, Golgi apparatus, and nucleus) in the ignition or avoidance of ferroptosis, while emphasizing their potential relevance for human disease and their targetability for pharmacological interventions.
Collapse
Affiliation(s)
- Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, The Third Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Daolin Tang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, The Third Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
242
|
Wang Z, Li M, Liu Y, Qiao Z, Bai T, Yang L, Liu B. Dihydroartemisinin triggers ferroptosis in primary liver cancer cells by promoting and unfolded protein response‑induced upregulation of CHAC1 expression. Oncol Rep 2021; 46:240. [PMID: 34558645 PMCID: PMC8485000 DOI: 10.3892/or.2021.8191] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/16/2021] [Indexed: 01/05/2023] Open
Abstract
Dihydroartemisinin (DHA), an artemisinin derivate, has been investigated as a potential antitumor drug in primary liver cancer (PLC). Ferroptosis is a form of iron-dependent cell death that can be driven by lipid peroxidation inducers. The present study aimed to determine whether and how DHA could promote the death of PLC cells by inducing ferroptosis. In total, four PLC cell lines with different p53 statuses, including Hep3B (p53 null), Huh7 (p53 mutant), PLC/PRF/5 (p53 mutant) and HepG2 (p53 wild-type), were treated with various concentrations of DHA. The effects of DHA on all three branches of the unfolded protein response (UPR) were evaluated. To deactivate the UPRs, small interfering RNA was used to knockdown the expression of activating transcription factor (ATF)4, X-box binding protein 1 (XBP1) or ATF6 in PLC cells. The effect of DHA on the promoter activity of Chac glutathione specific γ-glutamylcyclotransferase 1 (CHAC1) was evaluated using a dual luciferase reporter assay. The results revealed that DHA-induced death in PLC cells was irrelevant of the p53 status. PLC cells exposed to DHA displayed classic features of ferroptosis, such as increased lipid reactive oxygen species and malondialdehyde levels, an iron overload, and decreased activity or expression of glutathione (GSH), glutathione peroxidase 4, solute carrier family (SLC) 7 member 11 and SLC family 3 member 2. The antitumor effects of DHA in PLC cells were significantly weakened by two typical ferroptosis inhibitors, ferrostatin-1 and deferoxamine mesylate salt, whereas the antitumor effects were augmented following iron overload. Furthermore, DHA activated all three branches of the UPR (eukaryotic translation initiation factor 2 α kinase 3/eukaryotic translation initiation factor 2A/ATF4, inositol-requiring transmembrane kinase/endoribonuclease 1α/XBP1 and ATF6 branches) in vitro. Notably, DHA-induced ferroptosis was significantly attenuated following the knockdown of ATF4, XBP1 or ATF6 expression. In addition, the promoter activity of CHAC1, a gene capable of degrading GSH, was enhanced by DHA, but weakened when the aforementioned three UPR transcription factors were knocked down. In conclusion, the findings of the present study suggested that DHA may effectively induce ferroptosis in PLC cells through the activation of anti-survival UPRs and the upregulation of CHAC1 expression.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Mingxing Li
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yuanfeng Liu
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhentao Qiao
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Tao Bai
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ling Yang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Bo Liu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
243
|
Hu ZW, Wen YH, Ma RQ, Chen L, Zeng XL, Wen WP, Sun W. Ferroptosis Driver SOCS1 and Suppressor FTH1 Independently Correlate With M1 and M2 Macrophage Infiltration in Head and Neck Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:727762. [PMID: 34527677 PMCID: PMC8437260 DOI: 10.3389/fcell.2021.727762] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/11/2021] [Indexed: 12/23/2022] Open
Abstract
Objective To investigate the role of ferroptosis, an iron-dependent form of non-apoptotic cell death, in the head and neck squamous cell carcinoma (HNSCC) immune microenvironment. Materials and Methods A list of ferroptosis-related genes was obtained from the FerrDb database. Gene expression data were acquired from the cancer genome atlas (TCGA) and analyzed using the R language. Protein–protein interaction analysis was conducted using STRING and GeneMANIA. The correlations between gene expression levels and a patient’s survival were analyzed using GEPIA, the Kaplan–Meier estimate, and a multivariate Cox proportional hazards model. The expression results were verified using Oncomine and Human Protein Atlas data. We used the TIMER, GEPIA2, GEPIA2021, and TIMER2 databases to investigate the relationships between gene expression and infiltrating immune cells. Results Analysis of differentially expressed genes (DEGs) identified nine each ferroptosis drivers and ferroptosis suppressors, among which four genes correlated with survival as follows: two drivers (SOCS1, CDKN2A) associated with better survival and two suppressors (FTH1, CAV1) associated with poorer survival. Multivariate Cox survival analysis identified SOCS1 and FTH1 as independent prognostic factors for HNSCC, and their higher expression levels were verified using Oncomine and HPA data. The results acquired using TIMER, GEPIA2, GEPIA2021, and TIMER2 data revealed that the driver SOCS1 and the suppressor FTH1 independently correlated with M1 and M2 macrophage infiltration. Conclusions The ferroptosis driver SOCS1 and suppressor FTH1 are independent prognostic factors and that correlate with M1 and M2 macrophage infiltration in HNSCC. Targeting ferroptosis-immunomodulation may serve as a strategy to enhance the activity of immunotherapy.
Collapse
Affiliation(s)
- Zhang-Wei Hu
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Yi-Hui Wen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Ren-Qiang Ma
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Lin Chen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Xue-Lan Zeng
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Wei-Ping Wen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China.,Department of Otolaryngology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Sun
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
244
|
Lin HY, Ho HW, Chang YH, Wei CJ, Chu PY. The Evolving Role of Ferroptosis in Breast Cancer: Translational Implications Present and Future. Cancers (Basel) 2021; 13:cancers13184576. [PMID: 34572802 PMCID: PMC8466180 DOI: 10.3390/cancers13184576] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 01/03/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy among women worldwide. The discovery of regulated cell death processes has enabled advances in the treatment of BC. In the past decade, ferroptosis, a new form of iron-dependent regulated cell death caused by excessive lipid peroxidation has been implicated in the development and therapeutic responses of BC. Intriguingly, the induction of ferroptosis acts to suppress conventional therapy-resistant cells, and to potentiate the effects of immunotherapy. As such, pharmacological or genetic modulation targeting ferroptosis holds great potential for the treatment of drug-resistant cancers. In this review, we present a critical analysis of the current understanding of the molecular mechanisms and regulatory networks involved in ferroptosis, the potential physiological functions of ferroptosis in tumor suppression, its potential in therapeutic targeting, and explore recent advances in the development of therapeutic strategies for BC.
Collapse
Affiliation(s)
- Hung-Yu Lin
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan;
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan;
| | - Hui-Wen Ho
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan;
| | - Yen-Hsiang Chang
- Department of Nuclear Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chun-Jui Wei
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Correspondence: (C.-J.W.); (P.-Y.C.); Tel.: +886-97-5611-855 (P.-Y.C.)
| | - Pei-Yi Chu
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan;
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
- Correspondence: (C.-J.W.); (P.-Y.C.); Tel.: +886-97-5611-855 (P.-Y.C.)
| |
Collapse
|
245
|
Li Y, Wang X, Huang Z, Zhou Y, Xia J, Hu W, Wang X, Du J, Tong X, Wang Y. CISD3 inhibition drives cystine-deprivation induced ferroptosis. Cell Death Dis 2021; 12:839. [PMID: 34497268 PMCID: PMC8426496 DOI: 10.1038/s41419-021-04128-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/10/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022]
Abstract
Ferroptosis, a new form of programmed cell death, not only promotes the pathological process of various human diseases, but also regulates cancer progression. Current perspectives on the underlying mechanisms remain largely unknown. Herein, we report a member of the NEET protein family, CISD3, exerts a regulatory role in cancer progression and ferroptosis both in vivo and in vitro. Pan-cancer analysis from TCGA reveals that expression of CISD3 is generally elevated in various human cancers which are consequently associated with a higher hazard ratio and poorer overall survival. Moreover, knockdown of CISD3 significantly accelerates lipid peroxidation and accentuates free iron accumulation triggered by Xc- inhibition or cystine-deprivation, thus causing ferroptotic cell death. Conversely, ectopic expression of the shRNA-resistant form of CISD3 (CISD3res) efficiently ameliorates the ferroptotic cell death. Mechanistically, CISD3 depletion presents a metabolic reprogramming toward glutaminolysis, which is required for the fuel of mitochondrial oxidative phosphorylation. Both the inhibitors of glutaminolysis and the ETC process were capable of blocking the lipid peroxidation and ferroptotic cell death in the shCISD3 cells. Besides, genetic and pharmacological activation of mitophagy can rescue the CISD3 knockdown-induced ferroptosis by eliminating the damaged mitochondria. Noteworthily, GPX4 acts downstream of CISD3 mediated ferroptosis, which fails to reverse the homeostasis of mitochondria. Collectively, the present work provides novel insights into the regulatory role of CISD3 in ferroptotic cell death and presents a potential target for advanced antitumor activity through ferroptosis.
Collapse
Affiliation(s)
- Yanchun Li
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
- Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Xin Wang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Zhihui Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yi Zhou
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jun Xia
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wanye Hu
- Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Xu Wang
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jing Du
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Xiangmin Tong
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
- Bengbu Medical College, Bengbu, Anhui, 233000, China.
- Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Ying Wang
- Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
- Bengbu Medical College, Bengbu, Anhui, 233000, China.
- Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
246
|
Ren X, Li Y, Zhou Y, Hu W, Yang C, Jing Q, Zhou C, Wang X, Hu J, Wang L, Yang J, Wang H, Xu H, Li H, Tong X, Wang Y, Du J. Overcoming the compensatory elevation of NRF2 renders hepatocellular carcinoma cells more vulnerable to disulfiram/copper-induced ferroptosis. Redox Biol 2021; 46:102122. [PMID: 34482117 PMCID: PMC8416961 DOI: 10.1016/j.redox.2021.102122] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/28/2021] [Accepted: 08/28/2021] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the paramount causes of cancer-related death worldwide. Despite recent advances have been made in clinical treatments of HCC, the general prognosis of patients remains poor. Therefore, it is imperative to develop a less toxic and more effective therapeutic strategy. Currently, series of cellular, molecular, and pharmacological experimental approaches were utilized to address the unrecognized characteristics of disulfiram (DSF), pursuing the goal of repurposing DSF for cancer therapy. We found that DSF/Cu selectively exerted an efficient cytotoxic effect on HCC cell lines, and potently inhibited migration, invasion, and angiogenesis of HCC cells. Importantly, we confirmed that DSF/Cu could intensively impair mitochondrial homeostasis, increase free iron pool, enhance lipid peroxidation, and eventually result in ferroptotic cell death. Of note, a compensatory elevation of NRF2 accompanies the process of ferroptosis, and contributes to the resistance to DSF/Cu. Mechanically, we found that DSF/Cu dramatically activated the phosphorylation of p62, which facilitates competitive binding of Keap1, thus prolonging the half-life of NRF2. Notably, inhibition of NRF2 expression via RNA interference or pharmacological inhibitors significantly facilitated the accumulation of lipid peroxidation, and rendered HCC cells more sensitive to DSF/Cu induced ferroptosis. Conversely, fostering NRF2 expression was capable of ameliorating the cell death activated by DSF/Cu. Additionally, DSF/Cu could strengthen the cytotoxicity of sorafenib, and arrest tumor growth both in vitro and in vivo, by simultaneously inhibiting the signal pathway of NRF2 and MAPK kinase. In summary, these results provide experimental evidence that inhibition of the compensatory NRF2 elevation strengthens HCC cells more vulnerable to DSF/Cu induced ferroptosis, which facilitates the synergistic cytotoxicity of DSF/Cu and sorafenib.
Collapse
Affiliation(s)
- Xueying Ren
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yi Zhou
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wanye Hu
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Chen Yang
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Qiangan Jing
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Chaoting Zhou
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xu Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Jiayu Hu
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Luyang Wang
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jing Yang
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Hairui Wang
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Haifeng Xu
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huanjuan Li
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiangmin Tong
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Ying Wang
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China; Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
247
|
Yang J, Cao XH, Luan KF, Huang YD. Circular RNA FNDC3B Protects Oral Squamous Cell Carcinoma Cells From Ferroptosis and Contributes to the Malignant Progression by Regulating miR-520d-5p/SLC7A11 Axis. Front Oncol 2021; 11:672724. [PMID: 34434890 PMCID: PMC8382281 DOI: 10.3389/fonc.2021.672724] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common head and neck malignancy with increasing mortality and high recurrence. Ferroptosis is an emerging programed cell death and plays an essential role in tumorigenesis. Circular RNAs (circRNAs) have been reported as a type of critical regulators in OSCC development. In this study, we identified the function of circular RNA FNDC3B (circFNDC3B) in regulating ferroptosis during the malignant progression of OSCC. Our data demonstrated that the silencing of circFNDC3B by shRNA inhibited GPX4 and SLC7A11 expression and enhanced ROS, iron, and Fe2+ levels in OSCC cells. CircFNDC3B knockdown reinforced erastin-induced inhibitory effect on OSCC cells. The depletion of circFNDC3B repressed cell proliferation and enhanced cell apoptosis of OSCC cells. Mechanically, circFNDC3B was able to increase SLC7A11 by targeting miR-520d-5p. The overexpression of SLC7A11 reversed circFNDC3B depletion or miR-520d-5p-induced ferroptosis phenotypes of OSCC cells. Moreover, tumorgenicity assays in nude mice showed that the depletion of circFNDC3B repressed OSCC cell growth in vivo. Taken together, we concluded that circFNDC3B attenuated ferroptosis of OSCC cells and contributed to OSCC progression by regulating the miR-520d-5p/SLC7A11 axis. CircFNDC3B, miR-520d-5p, and SLC7A11 may serve as potential therapeutic targets of OSCC.
Collapse
Affiliation(s)
- Jie Yang
- Department of Stomatology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xing-Hua Cao
- Department of Stomatology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ke-Feng Luan
- Department of Stomatology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yun-Dong Huang
- Department of Stomatology, The Second People's Hospital of Weifang, Weifang, China
| |
Collapse
|
248
|
Zhao Y, Lu J, Mao A, Zhang R, Guan S. Autophagy Inhibition Plays a Protective Role in Ferroptosis Induced by Alcohol via the p62-Keap1-Nrf2 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:9671-9683. [PMID: 34388345 DOI: 10.1021/acs.jafc.1c03751] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Binge alcohol consumption is a serious health concern. Ferroptosis is an iron-dependent lipid peroxidation mediated cell death. Activation of the Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2) pathway has been shown to exert a protective effect by blunting the responses to ferroptosis inducers. The autophagy substrate p62 was demonstrated to modulate Nrf2 and contribute to the suppression of ferroptosis. Furthermore, autophagy inhibition resulted in the accumulation of p62, which is a specific substrate for this process. Therefore, we aimed to explore the protective effect of autophagy inhibition against alcohol-induced ferroptosis through activating the p62-Keap1-Nrf2 pathway. Our results demonstrated that alcohol induced ferroptosis, which could be significantly reduced by ferrostatin-1. Additionally, we found that autophagy inhibition could protect HepG2 cells against alcohol-induced ferroptosis by activating the p62-Keap1-Nrf2 pathway. Furthermore, inhibition of autophagy increased the expression of p62, which interacted with Keap1 to promote Nrf2 translocation into the nucleus and upregulation its target proteins ferritin heavy (FTH), ferroportin (FPN), and heme oxygenase-1 (HO-1). This study provides a theoretical basis for further elucidation of the relationship between autophagy and ferroptosis and lays a preliminary foundation for further research concerning dietary guidance in the prevention and treatment of diseases related to alcohol-induced ferroptosis.
Collapse
Affiliation(s)
- Yanan Zhao
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Ankang Mao
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Ranran Zhang
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| |
Collapse
|
249
|
Li Y, Xiao X, Wang H, Zhou Q, Jin Z, Zhang Y, Wang Y, Yue F, Zhou S, Yang J. Integrating network pharmacology and experimental models to investigate the mechanisms of dihydroartemisinin in preventing NSCLC progression via mTOR/HIF-1α signaling. Eur J Pharmacol 2021; 909:174411. [PMID: 34390710 DOI: 10.1016/j.ejphar.2021.174411] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/25/2022]
Abstract
Advanced Non-small cell lung cancer (NSCLC) is the most common type of lung cancer with a poor prognosis. The anti-malaria compounds dihydroartemisinin (DHA) have shown to regulate multiple targets and signaling pathways in cancers, but a global view of its mechanism of action remains elusive. In present study, we integrated network pharmacology and in vitro and in vivo experimental models to investigate the mechanisms of DHA in preventing NSCLC proliferation. We first proved that DHA inhibits the growth of lung cancer via inducing cell apoptosis and cell cycle arrest, then we integrated information from publicly available databases to predict interactions between DHA and its potential targets in NSCLC, as well as the signaling pathways involved. In this way we identified 118 common targets of DHA and NSCLC, and further analyzed with the correlation between these targets by KEGG and GO analysis. Our data indicate that mTOR/HIF-1α signaling is one of potential critical pathways involved in DHA-induced tumor inhibition in NSCLC. Finally, the data from human and mouse lung cancer cell lines and in mouse Lewis lung cancer models showed that DHA does decrease the expression level of mTOR and HIF-1α which supported the potential roles of mTOR/HIF-1α Signaling in NSCLC and deserves further investigation.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Artemisinins/pharmacology
- Artemisinins/therapeutic use
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- Disease Progression
- Drug Evaluation, Preclinical
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Mice
- Network Pharmacology
- Protein Interaction Maps/drug effects
- Protein Interaction Maps/genetics
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/metabolism
Collapse
Affiliation(s)
- Yanping Li
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiaoqian Xiao
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Huili Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Qi Zhou
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Zhao Jin
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yuxi Zhang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yi Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Fuping Yue
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Shiyi Zhou
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jiahui Yang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
250
|
Abstract
The major problems with cancer therapy are drug-induced side effects. There is an urgent need for safe anti-tumor drugs. Artemisinin is a Chinese herbal remedy for malaria with efficacy and safety. However, several studies reported that artemisinin causes neurotoxicity and cardiotoxicity in animal models. Recently, nanostructured drug delivery systems have been designed to improve therapeutic efficacy and reduce toxicity. Artemisinin has been reported to show anticancer properties. The anticancer effects of artemisinin appear to be mediated by inducing cell cycle arrest, promoting ferroptosis and autophagy, inhibiting cell metastasis. Therefore, the review is to concentrate on mechanisms and molecular targets of artemisinin as anti-tumor agents. We believe these will be important topics in realizing the potential of artemisinin and its derivatives as potent anticancer agents.
Collapse
Affiliation(s)
- Dongning Li
- Institute of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Jie Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyan Zhao
- Institute of Pharmaceutical Sciences, Southwest University, Chongqing, China
| |
Collapse
|