1
|
Liu Y, Stockwell BR, Jiang X, Gu W. p53-regulated non-apoptotic cell death pathways and their relevance in cancer and other diseases. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00842-3. [PMID: 40204927 DOI: 10.1038/s41580-025-00842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Programmed cell death is a mechanism that is crucial for numerous physiological and pathological processes. Whereas p53-mediated apoptosis is a major cell death pathway in cancer, accumulating evidence indicates that p53 also has crucial roles in controlling different non-apoptotic cell death (NACD) pathways, including ferroptosis, necroptosis, pyroptosis, autophagy-dependent cell death, entotic cell death, parthanatos and paraptosis, and may regulate PANoptosis, cuproptosis and disulfidptosis. Notably, the function of p53 in these NACDs substantially contributes to its biological effects, particularly in cancer development and other pathological processes. In this Review, we discuss recent advances in understanding the roles and underlying mechanisms of p53-mediated NACDs, focusing on ferroptosis, necroptosis and pyroptosis. We discuss the complex and distinct physiological settings in which NACDs are regulated by p53, and potential targeting of p53-regulated NACDs for the treatment of cancer and other human diseases. Finally, we highlight several important questions concerning p53-regulated NACDs that warrant further investigation.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, USA
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
2
|
Zheng J, Conrad M. Ferroptosis: when metabolism meets cell death. Physiol Rev 2025; 105:651-706. [PMID: 39661331 DOI: 10.1152/physrev.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
We present here a comprehensive update on recent advancements in the field of ferroptosis, with a particular emphasis on its metabolic underpinnings and physiological impacts. After briefly introducing landmark studies that have helped to shape the concept of ferroptosis as a distinct form of cell death, we critically evaluate the key metabolic determinants involved in its regulation. These include the metabolism of essential trace elements such as selenium and iron; amino acids such as cyst(e)ine, methionine, glutamine/glutamate, and tryptophan; and carbohydrates, covering glycolysis, the citric acid cycle, the electron transport chain, and the pentose phosphate pathway. We also delve into the mevalonate pathway and subsequent cholesterol biosynthesis, including intermediate metabolites like dimethylallyl pyrophosphate, squalene, coenzyme Q (CoQ), vitamin K, and 7-dehydrocholesterol, as well as fatty acid and phospholipid metabolism, including the biosynthesis and remodeling of ester and ether phospholipids and lipid peroxidation. Next, we highlight major ferroptosis surveillance systems, specifically the cyst(e)ine/glutathione/glutathione peroxidase 4 axis, the NAD(P)H/ferroptosis suppressor protein 1/CoQ/vitamin K system, and the guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin/dihydrofolate reductase axis. We also discuss other potential anti- and proferroptotic systems, including glutathione S-transferase P1, peroxiredoxin 6, dihydroorotate dehydrogenase, glycerol-3-phosphate dehydrogenase 2, vitamin K epoxide reductase complex subunit 1 like 1, nitric oxide, and acyl-CoA synthetase long-chain family member 4. Finally, we explore ferroptosis's physiological roles in aging, tumor suppression, and infection control, its pathological implications in tissue ischemia-reperfusion injury and neurodegeneration, and its potential therapeutic applications in cancer treatment. Existing drugs and compounds that may regulate ferroptosis in vivo are enumerated.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, Garching, Germany
| |
Collapse
|
3
|
Yu Q, Yang J, Chen H, Liu R, Hu R, Cai J, Yang S, Zheng B, Guo P, Cai Z, Zhang S, Zhang G. Macrophages hijack carbapenem-resistance hypervirulent Klebsiella pneumoniae by blocking SLC7A11/GSH-manipulated iron oxidative stress. Free Radic Biol Med 2025; 230:234-247. [PMID: 39965717 DOI: 10.1016/j.freeradbiomed.2025.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
Infection with carbapenem-resistant hypervirulent Klebsiella pneumoniae (CR-hvKP) is life-threatening because of its pronounced virulence and antibiotic resistance. Recent studies revealed that iron and ROS enhance the ability of macrophages to eliminate intracellular pathogenic bacteria. However, whether and how iron-related oxygen stress responses in macrophages elicit a protective role against CR-hvKP infection remains largely unknown. In a mouse model of CR-hvKP pulmonary infection, the production of the Solute Carrier Family 7 member 11 (SLC7A11) was increased. Treatment with the ferroptosis agonist Erastin or Sorafenib decreased the SLC7A11 expression and the bacterial load in infected lung tissues, alleviating CR-hvKP-induced acute lung injury, increasing the content of TLR4, ROS and LPO. In vitro experiments showed that CR-hvKP infection resulted in a remarkable time-dependent changes in the expression of SLC7A11, GSH, ferrous iron, ROS and LPO in MH-S cells. Mechanically, blocking the expression of SLC7A11 in CR-hvKP-infected MH-S cells increased iron and ROS, improving the ability of macrophages to clear CR-hvKP in an LPO-dependent manner. Taken together, our study reveals that improving iron-related oxygen stress via blocking the SLC7A11/GSH pathway promoting the macrophages to phagocytose and eliminate CR-hvKP, which provides a new promising strategy against CR-hvKP infection.
Collapse
Affiliation(s)
- Qing Yu
- Department of Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Jie Yang
- Department of Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China; Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, 325000, China
| | - Heyu Chen
- Department of Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Ruishan Liu
- Department of Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Ruomeng Hu
- Department of Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Jiachang Cai
- Clinical Microbiology Laboratory, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Shikuan Yang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Beiwen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Peng Guo
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310005, Zhejiang, China
| | - Zhijian Cai
- Institute of Immunology, And Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| | - Shufang Zhang
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, Hangzhou 310009, China.
| | - Gensheng Zhang
- Department of Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China; Key Laboratory of Multiple Organ Failure (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
4
|
Chen S, Hu S, Zhou Y, Cao J, Zhang H, Wang Y, Zhou J. Tick HRF-dependent ferroptosis pathway to promote tick acquisition of Babesia microti. Front Cell Infect Microbiol 2025; 15:1560152. [PMID: 40144593 PMCID: PMC11936993 DOI: 10.3389/fcimb.2025.1560152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
B. microti is a tick-transmitted zoonotic erythrocytic intracellular parasite. Ferroptosis is an iron-dependent form of programmed cell death that affects pathogen replication in the host. Currently, there is limited research concerning the effect of tick ferroptosis on Babesia infection and the underlying mechanism of action. The present study used a B. microti -mouse- Haemaphysalis longicornis infection model in which nymphs fed on the blood of B. microti-infected mice. The midgut divalent iron (p<0.01) and reactive oxygen species (ROS) (p<0.05) levels were significantly elevated in infected ticks, and transmission electron microscopy (TEM) showed that mitochondrial ridges were absent or decreased in size. Downregulation of ferritin 1 and glutathione peroxidase 4 (GPX4) in ticks infected with B. microti suggests that these changes promote ferroptosis. In vivo studies demonstrated that the ferroptosis promoter Erastin increased B. microti load (p<0.05), while the inhibitor Ferrostatin-1 effectively decreased load (p<0.01). Tick histamine-releasing factor (HRF), a protein related to the antioxidant system, was downregulated in infected nymphs compared with uninfected nymphs (p<0.05), and interference with HRF promoted tick acquisition of B. microti (p<0.001). Transcriptomic analyses showed that HRF interference promotes tick ferroptosis by downregulating ferritin 1 and GPX4. Meanwhile, interference with tick HRF molecules showed increased divalent iron and ROS and decreased mitochondrial ridges compared with controls. These findings highlight the critical role of tick HRF molecules in regulating ferroptosis and acquisition of B. microti, thereby providing important insights for a deeper understanding of the tick-Babesia interaction.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinlin Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural
Sciences, Shanghai, China
| |
Collapse
|
5
|
He Y, Lin Y, Song J, Song M, Nie X, Sun H, Xu C, Han Z, Cai J. From mechanisms to medicine: Ferroptosis as a Therapeutic target in liver disorders. Cell Commun Signal 2025; 23:125. [PMID: 40055721 PMCID: PMC11889974 DOI: 10.1186/s12964-025-02121-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/22/2025] [Indexed: 05/13/2025] Open
Abstract
In recent 10 years, ferroptosis has become a hot research direction in the scientific research community as a new way of cell death. Iron toxicity accumulation and lipotoxicity are unique features. Several studies have found that ferroptosis is involved in the regulation of the hepatic microenvironment and various hepatic metabolisms, thereby mediating the progression of related liver diseases. For example, NRF2 and FSP1, as important regulatory proteins of ferroptosis, are involved in the development of liver tumors and liver failure. In this manuscript, we present the mechanisms involved in ferroptosis, the concern of ferroptosis with the liver microenvironment and the progression of ferroptosis in various liver diseases. In addition, we summarize recent clinical advances in targeted ferroptosis therapy for related diseases. We expect that this manuscript can provide a new perspective for clinical treatment of related diseases.
Collapse
Affiliation(s)
- Yuqi He
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jinfeng Song
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Mingzhu Song
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Xiaoxia Nie
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Hong Sun
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Changyun Xu
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Zhongyu Han
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China.
| | - Juan Cai
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China.
| |
Collapse
|
6
|
Xiang L, Li Q, Guan Z, Wang G, Yu X, Zhang X, Zhang G, Hu J, Yang X, Li M, Bao X, Wang Y, Wang D. Oxyresveratrol as a novel ferroptosis inducer exhibits anticancer activity against breast cancer via the EGFR/PI3K/AKT/GPX4 signalling axis. Front Pharmacol 2025; 15:1527286. [PMID: 39881871 PMCID: PMC11775479 DOI: 10.3389/fphar.2024.1527286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Oxyresveratrol (ORes) exhibits significant anticancer activity, particularly against breast cancer. However, its exact mechanism of action (MOA) remains unclear. This study aimed to investigate the pharmacological activity and underlying MOA. Methods The inhibitory effect of ORes on breast cancer cell growth was confirmed, and the effective concentrations were determined for further experiments. Gene expression profiles (GEPs) were collected from MDA-MB-231 cells treated with ORes at varying concentrations using HTS2. Bioinformatics tools were used to predict the anticancer activity and MOA of ORes. Ferroptosis markers (ferrous ions, reactive oxygen species, lipid peroxidation, and GPX4 expression) were assessed, and mitochondrial morphology was observed. The effect of ORes on tumour growth was evaluated in vivo, along with the analysis of ferroptosis in tissues. The MOA was explored using L1000, Drug Gene DataBase (DGDB), and Western blotting analyses. Results ORes significantly reduces breast cancer cell viability and proliferation in a concentration-dependent manner, with IC50 values of 104.8 μM, 150.2 μM, and 143.6 μM in MDA-MB-231, BT-549, and 4T1 cells, respectively. GEPs induced by ORes were significantly enriched in the ferroptosis and PI3K/AKT signalling pathways. ORes inhibited breast cancer cell growth, increased intracellular ferrous ion levels, reactive oxygen species, and lipid peroxidation, and induced ferroptosis-related mitochondrial alterations. These effects were associated with decreased GPX4 expression and suppression of EGFR, phosphorylated PI3K, and phosphorylated AKT. ORes inhibited tumour growth, enhanced iron deposition, and reduced GPX4 expression in tumour tissues in vivo. Notably, treatment with the ferroptosis inhibitor ferrostatin-1 (Ferr-1) attenuated the anticancer effects of ORes, confirming the pivotal role of ferroptosis in ORes-mediated breast cancer inhibition. Conclusion ORes inhibits breast cancer cell growth by inducing ferroptosis through suppression of the EGFR/PI3K/AKT/GPX4 signalling axis. This study suggests that ORes holds promise as a potential therapeutic agent for breast cancer and warrants further investigation into its clinical applications and potential integration into existing treatment regimens.
Collapse
Affiliation(s)
- Lei Xiang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingzhou Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhiwei Guan
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guilin Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiankuo Yu
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianwen Zhang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guochen Zhang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jushan Hu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xue Yang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingrui Li
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xilinqiqige Bao
- Medical Innovation Center for Nationalities, Inner Mongolia Medical University, Hohhot, China
| | - Yumei Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
7
|
Wang H, Su Z, Qian Y, Shi B, Li H, An W, Xiao Y, Qiu C, Guo Z, Zhong J, Wu X, Chen J, Wang Y, Zeng W, Zhan L, Wang J. Pentraxin-3 modulates hepatocyte ferroptosis and the innate immune response in LPS-induced liver injury. MOLECULAR BIOMEDICINE 2024; 5:68. [PMID: 39666228 PMCID: PMC11638432 DOI: 10.1186/s43556-024-00227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
The liver plays a crucial role in the immune response during endotoxemia and is one of the critical targets for sepsis-related injuries. As a secretory factor involved in inflammation, pentraxin-3 (PTX3) has been demonstrated to regulate hepatic homeostasis; however, the relationship between PTX3 and cell crosstalk between immune cells and hepatocytes in the liver remains incompletely understood. In this study, we revealed that, compared with WT mice, Ptx3-/- mice with lipopolysaccharide (LPS)-induced endotoxemia exhibited alleviated liver damage, with reduced serum alanine transaminase and aspartate transaminase levels and an improved survival rate. Mechanistically, RNA-Seq and western blot results revealed that Ptx3 knockdown in hepatocytes increased the expression of Tfrc and Ccl20; consequently, Ptx3 deficiency regulated LPS-induced hepatocyte ferroptosis via increased mitochondrial reactive oxygen species and Fe2+ and recruited more macrophages by CCL20/CCR6 axis to be involved in inflammation and the clearance of harmful substances. Moreover, western blot and immunofluorescence staining confirmed that the NF-κB signaling pathway was upregulated upon LPS treatment in Ptx3-knockdown macrophages, promoting phagocytosis and polarization toward M1 macrophages. Collectively, our findings show that the absence of Ptx3 can ameliorate sepsis-induced liver injury by regulating hepatocyte ferroptosis and promote the recruitment and polarization of M1 macrophages. These findings offer a key basis for the development of effective treatments for acute infections.
Collapse
Affiliation(s)
- Huitong Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhaojie Su
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yunyun Qian
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Baojie Shi
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hao Li
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wenbin An
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yi Xiao
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Cheng Qiu
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhixiang Guo
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jianfa Zhong
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xia Wu
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jiajia Chen
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ying Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wei Zeng
- Department of Gastroenterology, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Linghui Zhan
- Department of Critical Care Medicine, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, 361004, China.
| | - Jie Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
8
|
Sui Y, Geng X, Wang Z, Zhang J, Yang Y, Meng Z. Targeting the regulation of iron homeostasis as a potential therapeutic strategy for nonalcoholic fatty liver disease. Metabolism 2024; 157:155953. [PMID: 38885833 DOI: 10.1016/j.metabol.2024.155953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/09/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024]
Abstract
With aging and the increasing incidence of obesity, nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide. NAFLD mainly includes simple hepatic steatosis, nonalcoholic steatohepatitis (NASH), liver fibrosis and hepatocellular carcinoma (HCC). An imbalance in hepatic iron homeostasis is usually associated with the progression of NAFLD and induces iron overload, reactive oxygen species (ROS) production, and lipid peroxide accumulation, which leads to ferroptosis. Ferroptosis is a unique type of programmed cell death (PCD) that is characterized by iron dependence, ROS production and lipid peroxidation. The ferroptosis inhibition systems involved in NAFLD include the solute carrier family 7 member 11 (SLC7A11)/glutathione (GSH)/glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1)/coenzyme Q10 (CoQ10)/nicotinamide adenine dinucleotide phosphate (NADPH) regulatory axes. The main promotion system involved is the acyl-CoA synthetase long-chain family (ACSL4)/arachidonic lipoxygenase 15 (ALOX15) axis. In recent years, an increasing number of studies have focused on the multiple roles of iron homeostasis imbalance and ferroptosis in the progression of NAFLD. This review highlights the latest studies about iron homeostasis imbalance- and ferroptosis-associated NAFLD, mainly including the physiology and pathophysiology of hepatic iron metabolism, hepatic iron homeostasis imbalance during the development of NAFLD, and key regulatory molecules and roles of hepatic ferroptosis in NAFLD. This review aims to provide innovative therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
- Yutong Sui
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China
| | - Xue Geng
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Ziwei Wang
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China
| | - Jing Zhang
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China
| | - Yanqun Yang
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China.
| | - Ziyu Meng
- NHC Key Laboratory of Hormones and Development, Tianjin Medical University Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
9
|
Dai Y, Liang Y, Liu C, Liu T, Chen L, Li Y. Can artemisinin and its derivatives treat malaria in a host-directed manner? Biochem Pharmacol 2024; 225:116260. [PMID: 38705539 DOI: 10.1016/j.bcp.2024.116260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Malaria is caused by an apicomplexan protozoan parasite, Plasmodium, and is transmitted through vectors. It remains a substantial health burden, especially in developing countries, leading to significant socioeconomic losses. Although the World Health Organization (WHO) has approved various antimalarial medications in the past two decades, the increasing resistance to these medications has worsened the situation. The development of drug resistance stems from genetic diversity among Plasmodium strains, impeding eradication efforts. Consequently, exploring innovative technologies and strategies for developing effective medications based on the host is crucial. Artemisinin and its derivatives (artemisinins) have been recommended by the WHO for treating malaria owing to their known effectiveness in killing the parasite. However, their potential to target the host for malaria treatment has not been investigated. This article concisely reviews the application of host-directed therapeutics, potential drug candidates targeting the host for treating malaria, and usage of artemisinins in numerous diseases. It underscores the importance of host-directed interventions for individuals susceptible to malaria, suggests the potential utility of artemisinins in host-directed malaria treatments, and posits that the modulation of host proteins with artemisinins may offer a means of intervening in host-parasite interactions. Further studies focusing on the host-targeting perspective of artemisinins can provide new insights into the mechanisms of artemisinin resistance and offer a unique opportunity for new antimalarial drug discovery.
Collapse
Affiliation(s)
- Yue Dai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yan Liang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chengcheng Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Tuo Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lina Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
10
|
Marques-da-Silva C, Schmidt-Silva C, Kurup SP. Hepatocytes and the art of killing Plasmodium softly. Trends Parasitol 2024; 40:466-476. [PMID: 38714463 PMCID: PMC11156546 DOI: 10.1016/j.pt.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 05/09/2024]
Abstract
The Plasmodium parasites that cause malaria undergo asymptomatic development in the parenchymal cells of the liver, the hepatocytes, prior to infecting erythrocytes and causing clinical disease. Traditionally, hepatocytes have been perceived as passive bystanders that allow hepatotropic pathogens such as Plasmodium to develop relatively unchallenged. However, now there is emerging evidence suggesting that hepatocytes can mount robust cell-autonomous immune responses that target Plasmodium, limiting its progression to the blood and reducing the incidence and severity of clinical malaria. Here we discuss our current understanding of hepatocyte cell-intrinsic immune responses that target Plasmodium and how these pathways impact malaria.
Collapse
Affiliation(s)
- Camila Marques-da-Silva
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Clyde Schmidt-Silva
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Samarchith P Kurup
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA.
| |
Collapse
|
11
|
Li J, Yang J, Xian Q, Su H, Ni Y, Wang L. Kaempferitrin attenuates unilateral ureteral obstruction-induced renal inflammation and fibrosis in mice by inhibiting NOX4-mediated tubular ferroptosis. Phytother Res 2024; 38:2656-2668. [PMID: 38487990 DOI: 10.1002/ptr.8191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 06/13/2024]
Abstract
Tubular ferroptosis significantly contributes to renal inflammation and fibrosis, critical factors in chronic kidney disease (CKD). This study aims to investigate Kaempferitrin, a potent flavonoid glycoside from Bauhinia forficata leaves, renowned for its anti-inflammatory and antitumor effects, and to elucidate its potential mechanisms in mitigating inflammation and fibrosis induced by tubular ferroptosis. The study investigated Kaempferitrin's impact on tubular ferroptosis using a unilateral ureteral obstruction (UUO) model-induced renal inflammation and fibrosis. In vitro, erastin-induced ferroptosis in primary tubular epithelial cells (TECs) was utilized to further explore Kaempferitrin's effects. Additionally, NADPH oxidase 4 (NOX4) transfection in TECs and cellular thermal shift assay (CETSA) were conducted to identify Kaempferitrin's target protein. Kaempferitrin effectively improved renal function, indicated by reduced serum creatinine and blood urea nitrogen levels. In the UUO model, it significantly reduced tubular necrosis, inflammation, and fibrosis. Its renoprotective effects were linked to ferroptosis inhibition, evidenced by decreased iron, 4-hydroxynonenal (4-HNE), and malondialdehyde (MDA) levels, and increased glutathione (GSH). Kaempferitrin also normalized glutathione peroxidase 4 (GPX4) and Solute Carrier Family 7 Member 11(SLC7A11) expression, critical ferroptosis mediators. In vitro, it protected TECs from ferroptosis and consistently suppressed NOX4 expression. NOX4 transfection negated Kaempferitrin's antiferroptosis effects, while CETSA confirmed Kaempferitrin-NOX4 interaction. Kaempferitrin shows promise as a nephroprotective agent by inhibiting NOX4-mediated ferroptosis in tubular cells, offering potential therapeutic value for CKD.
Collapse
Affiliation(s)
- Jianchun Li
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jieke Yang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Qianwen Xian
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hongwei Su
- Department of Urology, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yufang Ni
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Li Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Yang B, Shu W, Hu J, Wang Z, Wu J, Su J, Tan J, Yu B, Zhang X. Aberrant Expression of SLC7A11 Impairs the Antimicrobial Activities of Macrophages in Staphylococcus Aureus Osteomyelitis in Mice. Int J Biol Sci 2024; 20:2555-2575. [PMID: 38725861 PMCID: PMC11077379 DOI: 10.7150/ijbs.93592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Staphylococcus aureus (S. aureus) persistence in macrophages, potentially a reservoir for recurrence of chronic osteomyelitis, contributes to resistance and failure in treatment. As the mechanisms underlying survival of S. aureus in macrophages remain largely unknown, there has been no treatment approved. Here, in a mouse model of S. aureus osteomyelitis, we identified significantly up-regulated expression of SLC7A11 in both transcriptomes and translatomes of CD11b+F4/80+ macrophages, and validated a predominant distribution of SLC7A11 in F4/80+ cells around the S. aureus abscess. Importantly, pharmacological inhibition or genetic knockout of SLC7A11 promoted the bactericidal function of macrophages, reduced bacterial burden in the bone and improved bone structure in mice with S. aureus osteomyelitis. Mechanistically, aberrantly expressed SLC7A11 down-regulated the level of intracellular ROS and reduced lipid peroxidation, contributing to the impaired bactericidal function of macrophages. Interestingly, blocking SLC7A11 further activated expression of PD-L1 via the ROS-NF-κB axis, and a combination therapy of targeting both SLC7A11 and PD-L1 significantly enhanced the efficacy of clearing S. aureus in vitro and in vivo. Our findings suggest that targeting both SLC7A11 and PD-L1 is a promising therapeutic approach to reprogram the bactericidal function of macrophages and promote bacterial clearance in S. aureus osteomyelitis.
Collapse
Affiliation(s)
- Bingsheng Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wen Shu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Department of Trauma Orthopedics, Liuzhou People's Hospital, Liuzhou, Guangxi, China
| | - Jin Hu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhongwen Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jichang Wu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jianwen Su
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jianye Tan
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xianrong Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
13
|
Adegboro AG, Afolabi IS. Molecular mechanisms of mitochondria-mediated ferroptosis: a potential target for antimalarial interventions. Front Cell Dev Biol 2024; 12:1374735. [PMID: 38660623 PMCID: PMC11039840 DOI: 10.3389/fcell.2024.1374735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Ferroptosis is an iron-dependent form of regulated cell death characterized by glutathione (GSH) depletion, glutathione peroxidase 4 (GPX4) inactivation, and the build-up of lipotoxic reactive species. Ferroptosis-targeted induction is a promising therapeutic approach for addressing antimalarial drug resistance. In addition to being the primary source of intracellular energy supply and reactive oxygen species (ROS) generation, mitochondria actively participate in diverse forms of regulated cell death, including ferroptosis. Altered mitochondrial morphology and functionality are attributed to ferroptosis. Diverse mitochondria-related proteins and metabolic activities have been implicated in fine-tuning the action of ferroptosis inducers. Herein, we review recent progress in this evolving field, elucidating the numerous mechanisms by which mitochondria regulate ferroptosis and giving an insight into the role of the organelle in ferroptosis. Additionally, we present an overview of how mitochondria contribute to ferroptosis in malaria. Furthermore, we attempt to shed light on an inclusive perspective on how targeting malaria parasites' mitochondrion and attacking redox homeostasis is anticipated to induce ferroptosis-mediated antiparasitic effects.
Collapse
Affiliation(s)
- Adegbolagun Grace Adegboro
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Israel Sunmola Afolabi
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
| |
Collapse
|
14
|
Tang D, Kroemer G, Kang R. Ferroptosis in immunostimulation and immunosuppression. Immunol Rev 2024; 321:199-210. [PMID: 37424139 DOI: 10.1111/imr.13235] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/11/2023]
Abstract
Ferroptosis is a form of iron-dependent regulated cell death characterized by the accumulation of toxic lipid peroxides, particularly in the plasma membrane, leading to lytic cell death. While it plays a crucial role in maintaining the overall health and proper functioning of multicellular organisms, it can also contribute to tissue damage and pathological conditions. Although ferroptotic damage is generally recognized as an immunostimulatory process associated with the release of damage-associated molecular patterns (DAMPs), the occurrence of ferroptosis in immune cells or the release of immunosuppressive molecules can result in immune tolerance. Consequently, there is ongoing exploration of targeting the upstream signals or the machinery of ferroptosis to therapeutically enhance or inhibit the immune response. In addition to introducing the core molecular mechanisms of ferroptosis, we will focus on the immune characteristics of ferroptosis in pathological conditions, particularly in the context of infection, sterile inflammation, and tumor immunity.
Collapse
Affiliation(s)
- Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
15
|
Hu Y, He B, Cao Q, Li Y, Tang Y, Cao T, Peng B, Zhou X, Liu S. Crosstalk of ferroptosis and oxidative stress in infectious diseases. Front Mol Biosci 2023; 10:1315935. [PMID: 38131014 PMCID: PMC10733455 DOI: 10.3389/fmolb.2023.1315935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Ferroptosis is a type of programmed cell death that pathogens can leverage to enhance their replication, transmission, and pathogenicity. Hosts typically combat pathogenic infections by utilizing oxidative stress as a defense mechanism. Nonetheless, some pathogens can trigger considerable oxidative stress while infecting, inducing an intense inflammatory response in the host's immune system and activating cell death. The process of ferroptosis is closely linked to oxidative stress, with their interaction exerting a substantial impact on the outcome of infectious diseases. This article presents an overview of the interrelated mechanisms of both Ferroptosis and oxidative stress in infectious diseases, identifying potential targets for treating such diseases in the context of their interaction.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Shuangquan Liu
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
16
|
Glennon EK, Wei L, Roobsoong W, Primavera VI, Tongogara T, Yee CB, Sattabongkot J, Kaushansky A. Host kinase regulation of Plasmodium vivax dormant and replicating liver stages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566868. [PMID: 38014051 PMCID: PMC10680662 DOI: 10.1101/2023.11.13.566868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Upon transmission to the liver, Plasmodium vivax parasites form replicating schizonts, which continue to initiate blood-stage infection, or dormant hypnozoites that reactivate weeks to months after initial infection. P. vivax phenotypes in the field vary significantly, including the ratio of schizonts to hypnozoites formed and the frequency and timing of relapse. Evidence suggests that both parasite genetics and environmental factors underly this heterogeneity. We previously demonstrated that data on the effect of a panel of kinase inhibitors with overlapping targets on Plasmodium liver stage infection, in combination with a computational approach called kinase regression (KiR), can be used to uncover novel host regulators of infection. Here, we applied KiR to evaluate the extent to which P. vivax liver-stage parasites are susceptible to changes in host kinase activity. We identified a role for a subset of host kinases in regulating schizont and hypnozoite infection and schizont size and characterized overlap as well as variability in host phosphosignaling dependencies between parasite forms and across multiple patient isolates. Striking, our data point to variability in host dependencies across P. vivax isolates, suggesting one possible origin of the heterogeneity observed across P. vivax in the field.
Collapse
|
17
|
Douradinha B. Does attenuated plasmodial sporozoite-mediated protection require peroxynitrite? Trends Parasitol 2023; 39:808-811. [PMID: 37574429 DOI: 10.1016/j.pt.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/03/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023]
Abstract
Attenuated plasmodial sporozoite-induced immune response includes intrahepatic nitric oxide (NO) production, which promotes apoptosis of infected hepatocytes and consequent parasite clearance. NO in excess reacts with superoxide, forming peroxynitrite, a powerful cytotoxic agent. Here, I suggest that peroxynitrite proapoptotic action may contribute to the attenuated malarial sporozoite-mediated protection.
Collapse
Affiliation(s)
- Bruno Douradinha
- Nykode Therapeutics ASA, Oslo Science Park, Gaustadalléen 21, 0349 Oslo, Norway.
| |
Collapse
|
18
|
Li Q, Chen K, Zhang T, Jiang D, Chen L, Jiang J, Zhang C, Li S. Understanding sorafenib-induced ferroptosis and resistance mechanisms: Implications for cancer therapy. Eur J Pharmacol 2023; 955:175913. [PMID: 37460053 DOI: 10.1016/j.ejphar.2023.175913] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/20/2023] [Accepted: 07/13/2023] [Indexed: 07/22/2023]
Abstract
Sorafenib is an important first-line treatment option for liver cancer due to its well-characterized safety profile. While novel first-line drugs may have better efficacy than Sorafenib, they also have limitations such as worse safety and cost-effectiveness. In addition to inducing apoptosis, Sorafenib can also trigger ferroptosis, which has recently been recognized as an immunogenic cell death, unleashing new possibilities for cancer treatment. However, resistance to Sorafenib-induced ferroptosis remains a major challenge. To overcome this resistance and augment the efficacy of Sorafenib, a wide range of nanomedicines has been developed to amplify its pro-ferroptotic effects. This review highlights the mechanisms underlying Sorafenib-triggered ferroptosis and its resistance, and outlines innovative strategies, particularly nanomedicines, to overcome ferroptosis resistance. Moreover, we summarize molecular biomarkers that signify resistance to Sorafenib-mediated ferroptosis, which can assist in predicting therapeutic outcomes.
Collapse
Affiliation(s)
- Qiuhong Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Kexin Chen
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Tianyi Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Donghui Jiang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
| | - Chunxiang Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Shengbiao Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
19
|
Cai H, Luo S, Liu Q, Zhou Q, Yan Z, Kang Z, Liao S, Li J, Lv M, Lin X, Hu J, Yu S, Zhang J, Qi N, Sun M. Effects of a complex probiotic preparation, Fengqiang Shengtai and coccidiosis vaccine on the performance and intestinal microbiota of broilers challenged with Eimeria spp. Parasit Vectors 2023; 16:253. [PMID: 37501177 PMCID: PMC10375739 DOI: 10.1186/s13071-023-05855-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Coccidiosis, a prominent intestinal protozoan disease, carries significant economic implications for the poultry industry. The aim of this study was to evaluate the effects of Fengqiang Shengtai (BLES), a probiotics product, and coccidiosis vaccine in modulating the intestinal microbiome and providing insight into mitigating the occurrence and management of avian coccidiosis. METHODS Broilers included in the study were divided into four pre-treatment groups: the Pre-Con group (commercial diet), Pre-BLES group (BLES supplement), Pre-Vac group (coccidiosis vaccination) and Pre-Vac-BLES group (combined vaccination and BLES). Body weight gain, feed consumption and feed conversion ratio were monitored from age 25 to 55 days. Cecum contents were collected at 8 and 15 days of age for comparative analysis of intestinal microbiomes. In the Pre-BLES and Pre-Vac-BLES groups, probiotics were administered at a dose of 0.01 g per chicken between ages 3 to 6 days and 10-13 days. At 3 days of age, chickens in the Pre-Vac and Pre-Vac-BLES groups were vaccinated with 1700 sporulated oocysts of the live coccidiosis vaccine per chicken. At the age of 25 days, Eimeria spp. challenge experiments were performed based on the aforementioned immunization strategy, and the oocysts per gram (OPG) in the feces, intestinal lesion score and intestinal pathological characteristics were evaluated. Specifically, 30 chickens were randomly selected from each group and orally administered 34,000 sporulated oocysts of Eimeria spp. per chicken, re-defined as Eimeria group, BLES-Eimeria group, Vac-Eimeria group and Vac-BLES-Eimeria group, respectively. Additionally, 30 chickens were randomly selected from the Pre-Con group and included as negative control without Eimeria spp. CHALLENGE Intestinal microbiota was sequenced and analyzed when the broilers were 32 days old. RESULTS A significant improvement was observed in body weight gain of the broilers in the Pre-BLES and Pre-Vac-BLES group at 45 days of age. Analysis of the intestinal microbiota revealed a positive correlation between the experimental groups receiving BLES and coccidiosis vaccines at 8 and 15 days of age with the Enterococcus genus and Lachnospiraceae NK4A136 group, respectively. In addition to the reduced lesion score and OPG values, the combination of coccidiosis vaccine and BLES also reduced the intestinal epithelial abscission induced by coccidiosis vaccines. The results of intestinal microbial function prediction demonstrated that N-glycan biosynthesis and ferroptosis were the prominent signal pathways in the Vac-BLES-Eimeria group. CONCLUSIONS Taken together, the results of the present study suggest that supplementation of BLES with coccidiosis vaccine represents a promising strategy for improving growth performance, alleviating clinical manifestations and inducing favorable alterations to the intestinal microbiota in broiler chickens affected by coccidiosis.
Collapse
Affiliation(s)
- Haiming Cai
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China
| | - Shengjun Luo
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China
| | - Qihong Liu
- Jiangsu HFQ Biotechnology Co., Ltd., Haimen, Jiangsu Province, People's Republic of China
| | - Qingfeng Zhou
- Wen's Group Academy, Wen's Foodstuffs Group Co., Ltd., Xinxing, Guangdong, 527400, People's Republic of China
| | - Zhuanqiang Yan
- Wen's Group Academy, Wen's Foodstuffs Group Co., Ltd., Xinxing, Guangdong, 527400, People's Republic of China
| | - Zhen Kang
- Qingdao Vland Biotech Group Co., Ltd., Qingdao, Shandong Province, People's Republic of China
| | - Shenquan Liao
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China
| | - Juan Li
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China
| | - Minna Lv
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China
| | - Xuhui Lin
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China
| | - Junjing Hu
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China
| | - Shuilan Yu
- Wen's Group Academy, Wen's Foodstuffs Group Co., Ltd., Xinxing, Guangdong, 527400, People's Republic of China
| | - Jianfei Zhang
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China
| | - Nanshan Qi
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China.
- Laboratory of Parasitology, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China.
| | - Mingfei Sun
- Zhaoqing/Maoming Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China.
- Laboratory of Parasitology, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Jinying Road, Tianhe District, Guangzhou, 510640, Guangdong, People's Republic of China.
| |
Collapse
|
20
|
Wang H, Qiao C, Guan Q, Wei M, Li Z. Nanoparticle-mediated synergistic anticancer effect of ferroptosis and photodynamic therapy: Novel insights and perspectives. Asian J Pharm Sci 2023; 18:100829. [PMID: 37588992 PMCID: PMC10425855 DOI: 10.1016/j.ajps.2023.100829] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/05/2023] [Accepted: 07/02/2023] [Indexed: 08/18/2023] Open
Abstract
Current antitumor monotherapy has many limitations, highlighting the need for novel synergistic anticancer strategies. Ferroptosis is an iron-dependent form of nonapoptotic cell death that plays a pivotal regulatory role in tumorigenesis and treatment. Photodynamic therapy (PDT) causes irreversible chemical damage to target lesions and is widely used in antitumor therapy. However, PDT's effectiveness is usually hindered by several obstacles, such as hypoxia, excess glutathione (GSH), and tumor resistance. Ferroptosis improves the anticancer efficacy of PDT by increasing oxygen and reactive oxygen species (ROS) or reducing GSH levels, and PDT also enhances ferroptosis induction due to the ROS effect in the tumor microenvironment (TME). Strategies based on nanoparticles (NPs) can subtly exploit the potential synergy of ferroptosis and PDT. This review explores recent advances and current challenges in the landscape of the underlying mechanisms regulating ferroptosis and PDT, as well as nano delivery system-mediated synergistic anticancer activity. These include polymers, biomimetic materials, metal organic frameworks (MOFs), inorganics, and carrier-free NPs. Finally, we highlight future perspectives of this novel emerging paradigm in targeted cancer therapies.
Collapse
Affiliation(s)
- Haiying Wang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Chu Qiao
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qiutong Guan
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Zhenhua Li
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
21
|
Gao N, Tang AL, Liu XY, Chen J, Zhang GQ. p53-Dependent ferroptosis pathways in sepsis. Int Immunopharmacol 2023; 118:110083. [PMID: 37028271 DOI: 10.1016/j.intimp.2023.110083] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 04/09/2023]
Abstract
Sepsis is caused by complex infections, trauma, and major surgery that results in high morbidity and mortality. As one of the leading causes of death in the intensive care unit (ICU), sepsis causes organ dysfunction and death via a vicious cycle of uncontrolled inflammatory responses and immunosuppression. Ferroptosis is an iron-dependent cellular death pathway driven by the accumulation of lipid peroxides, which occurs in sepsis. p53 is an important regulator of ferroptosis. Under intracellular/extracellular stimulation and pressure, p53 acts as a transcription factor to regulate the expression of downstream genes, which help cells/bodies to resist stimuli. p53 can also function independently as an important mediator. The understanding of key cellular and molecular mechanisms of ferroptosis facilitates the prognosis of sepsis. This article describes the molecular mechanism and role of p53 in sepsis-induced ferroptosis, and introduces some potential therapeutic targets for sepsis-induced ferroptosis, which highlights the dominant and potential therapeutic role of p53 in sepsis. Keywords: p53, acetylation, Sirt3, ferroptosis, sepsis, therapy.
Collapse
Affiliation(s)
- Nan Gao
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Emergency, China-Japan Friendship Hospital, No. 2 Yinghua Dongjie, Beijing 100029, China
| | - A-Ling Tang
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiao-Yu Liu
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Emergency, China-Japan Friendship Hospital, No. 2 Yinghua Dongjie, Beijing 100029, China
| | - Jie Chen
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Emergency, China-Japan Friendship Hospital, No. 2 Yinghua Dongjie, Beijing 100029, China
| | - Guo-Qiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, No. 2 Yinghua Dongjie, Beijing 100029, China.
| |
Collapse
|
22
|
Wagner MP, Chitnis CE. Lipid peroxidation and its repair in malaria parasites. Trends Parasitol 2023; 39:200-211. [PMID: 36642689 DOI: 10.1016/j.pt.2022.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023]
Abstract
During its life cycle, the human malaria parasite Plasmodium falciparum is subjected to elevated levels of oxidative stress that cause damage to membrane lipids, a process referred to as lipid peroxidation. Control and repair of lipid peroxidation is critical for survival of P. falciparum. Here, we present an introduction into lipid peroxidation and review the current knowledge about the control and repair of the damage caused by lipid peroxidation in P. falciparum blood stages. We also review the recent identification of host peroxiredoxin 6 (PRDX6), as a key lipid-peroxidation-repair enzyme in P. falciparum blood stages. Such critical host factors provide novel targets for development of drugs against malaria.
Collapse
Affiliation(s)
- Matthias Paulus Wagner
- Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, Paris, France
| | - Chetan E Chitnis
- Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, Paris, France.
| |
Collapse
|
23
|
Tong Q, Qin W, Li Z, Liu C, Wang Z, Chu Y, Xu X. SLC12A5 promotes hepatocellular carcinoma growth and ferroptosis resistance by inducing ER stress and cystine transport changes. Cancer Med 2023; 12:8526-8541. [PMID: 36645171 PMCID: PMC10134347 DOI: 10.1002/cam4.5605] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/10/2022] [Accepted: 12/21/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) has a poor prognosis and new effective treatments are needed. SLC12A5 plays important roles in multiple complex pathological states and is overexpressed in a variety of malignancies. However, the effects of SLC12A5 in HCC have not been determined. METHODS SLC12A5 expression was assessed by immunostaining and western blotting. A cell viability assay was used to detect cell proliferation. Flow cytometry was used to evaluate the intracellular calcium concentration and cell cycle. Ferroptosis was detected by transmission electron microscopy, lipid peroxidation, and glutathione assays. Subcutaneous tumor formation experiments were used to validate the tumorigenic effect of SLC12A5 in vivo. RNA-seq was used to evaluate the molecular mechanisms underlying the effects of SLC12A5. The therapeutic efficacy of targeting SLC12A5 was assessed in a patient-derived xenograft (PDX) model. RESULTS High SLC12A5 expression was strongly associated with a poor clinical prognosis and promoted HCC growth. Mechanistically, SLC12A5 promoted ER stress to enhance calcium release and upregulated PNCK expression levels. Concomitantly, PNCK was significantly activated by calcium ions released from the ER. PNCK activated and induced the phosphorylation of PI3K/AKT/mTOR pathway components. Furthermore, SLC12A5 inhibited ferroptosis in HCC by upregulating the expression of xCT, a cystine transporter. CONCLUSION High SLC12A5 levels were correlated with a poor prognosis, promoted tumorigenesis, and inhibited ferroptosis in HCC. These findings suggested that SLC12A5 is a therapeutic target and provide insight into the link between ER stress and ferroptosis in HCC.
Collapse
Affiliation(s)
- Qing Tong
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato‐Biliary‐Pancreatic Surgery, Department of SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
- Department of Hepato‐Biliary‐Pancreatic SurgeryThe 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital)UrumqiChina
| | - Wei Qin
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato‐Biliary‐Pancreatic Surgery, Department of SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Zheng‐Hao Li
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato‐Biliary‐Pancreatic Surgery, Department of SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Chun Liu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato‐Biliary‐Pancreatic Surgery, Department of SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Zi‐Cheng Wang
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato‐Biliary‐Pancreatic Surgery, Department of SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Yuan Chu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato‐Biliary‐Pancreatic Surgery, Department of SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Xun‐Di Xu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato‐Biliary‐Pancreatic Surgery, Department of SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
- Department of General SurgeryThe South China Hospital of Shenzhen UniversityShenzhenChina
| |
Collapse
|
24
|
Chandley P, Ranjan R, Kumar S, Rohatgi S. Host-parasite interactions during Plasmodium infection: Implications for immunotherapies. Front Immunol 2023; 13:1091961. [PMID: 36685595 PMCID: PMC9845897 DOI: 10.3389/fimmu.2022.1091961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Malaria is a global infectious disease that remains a leading cause of morbidity and mortality in the developing world. Multiple environmental and host and parasite factors govern the clinical outcomes of malaria. The host immune response against the Plasmodium parasite is heterogenous and stage-specific both in the human host and mosquito vector. The Plasmodium parasite virulence is predominantly associated with its ability to evade the host's immune response. Despite the availability of drug-based therapies, Plasmodium parasites can acquire drug resistance due to high antigenic variations and allelic polymorphisms. The lack of licensed vaccines against Plasmodium infection necessitates the development of effective, safe and successful therapeutics. To design an effective vaccine, it is important to study the immune evasion strategies and stage-specific Plasmodium proteins, which are targets of the host immune response. This review provides an overview of the host immune defense mechanisms and parasite immune evasion strategies during Plasmodium infection. Furthermore, we also summarize and discuss the current progress in various anti-malarial vaccine approaches, along with antibody-based therapy involving monoclonal antibodies, and research advancements in host-directed therapy, which can together open new avenues for developing novel immunotherapies against malaria infection and transmission.
Collapse
Affiliation(s)
- Pankaj Chandley
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Ravikant Ranjan
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Soma Rohatgi
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India,*Correspondence: Soma Rohatgi,
| |
Collapse
|
25
|
Lin S, Jin X, Gu H, Bi F. Relationships of ferroptosis-related genes with the pathogenesis in polycystic ovary syndrome. Front Med (Lausanne) 2023; 10:1120693. [PMID: 36873892 PMCID: PMC9981782 DOI: 10.3389/fmed.2023.1120693] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023] Open
Abstract
Background Numerous studies have suggested that ferroptosis plays a significant role in the development of polycystic ovary syndrome (PCOS), but the mechanism remains unclear. Methods In this study, we explored the role of ferroptosis-related genes in the pathogenesis of PCOS using a comprehensive bioinformatics method. First, we downloaded several Gene Expression Omnibus (GEO) datasets and combined them into a meta-GEO dataset. Differential expression analysis was performed to screen for significant ferroptosis-related genes between the normal and PCOS samples. The least absolute shrinkage selection operator regression and support vector machine-recursive feature elimination were used to select the best signs to construct a PCOS diagnostic model. Receiver operating characteristic curve analysis and decision curve analysis were applied to test the performance of the model. Finally, a ceRNA network-related ferroptosis gene was constructed. Results Five genes, namely, NOX1, ACVR1B, PHF21A, FTL, and GALNT14, were identified from 10 differentially expressed ferroptosis-related genes to construct a PCOS diagnostic model. Finally, a ceRNA network including 117 lncRNAs, 67 miRNAs, and five ferroptosis-related genes was constructed. Conclusion Our study identified five ferroptosis-related genes that may be involved in the pathogenesis of PCOS, which may provide a novel perspective for the clinical diagnosis and treatment of PCOS.
Collapse
Affiliation(s)
- Shuang Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xin Jin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Gu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fangfang Bi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
26
|
Eleutheroside B ameliorated high altitude pulmonary edema by attenuating ferroptosis and necroptosis through Nrf2-antioxidant response signaling. Biomed Pharmacother 2022; 156:113982. [DOI: 10.1016/j.biopha.2022.113982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
|
27
|
Bagayoko S, Meunier E. Emerging roles of ferroptosis in infectious diseases. FEBS J 2022; 289:7869-7890. [PMID: 34670020 DOI: 10.1111/febs.16244] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/06/2021] [Accepted: 10/20/2021] [Indexed: 01/14/2023]
Abstract
In living organisms, lipid peroxidation is a continuously occurring cellular process and therefore involved in various physiological and pathological contexts. Among the broad variety of lipids, polyunsaturated fatty acids (PUFA) constitute a major target of oxygenation either when released as mediators by phospholipases or when present in membranous phospholipids. The last decade has seen the characterization of an iron- and lipid peroxidation-dependent cell necrosis, namely, ferroptosis, that involves the accumulation of peroxidized PUFA-containing phospholipids. Further studies could link ferroptosis in a very large body of (physio)-pathological processes, including cancer, neurodegenerative, and metabolic diseases. In this review, we mostly focus on the emerging involvement of lipid peroxidation-driven ferroptosis in infectious diseases, and the immune consequences. We also discuss the putative ability of microbial virulence factors to exploit or to dampen ferroptosis regulatory pathways to their own benefit.
Collapse
Affiliation(s)
- Salimata Bagayoko
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, France
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, France
| |
Collapse
|
28
|
Zeng C, Lin J, Zhang K, Ou H, Shen K, Liu Q, Wei Z, Dong X, Zeng X, Zeng L, Wang W, Yao J. SHARPIN promotes cell proliferation of cholangiocarcinoma and inhibits ferroptosis via p53/SLC7A11/GPX4 signaling. Cancer Sci 2022; 113:3766-3775. [PMID: 35968603 PMCID: PMC9633309 DOI: 10.1111/cas.15531] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 07/18/2022] [Accepted: 08/05/2022] [Indexed: 11/27/2022] Open
Abstract
SHARPIN is a tumor-associated gene involved in the growth and proliferation of many tumor types. A function of SHARPIN in cholangiocarcinoma (CCA) is so far unclear. Here, we studied the role and function of SHARPIN in CCA and revealed its relevant molecular mechanism. The expression of SHARPIN was analyzed in cholangiocarcinoma tissues from patients using immunohistochemistry, quantitative PCR, and western blot analysis. Expression of SHARPIN was suppressed/overexpressed by siRNA silencing or lentiviral overexpression vector, and the effect on cell proliferation was determined by the CCK-8 assay and flow cytometry. Accumulation of reactive oxygen species was measured with MitoTracker, and JC-1 staining showed mitochondrial fission/fusion and mitochondrial membrane potential changes as a result of the silencing or overexpression. The ferroptosis marker solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), and the antioxidant enzymes superoxide dismutase 1 (SOD-1) and SOD-2 were analyzed by western blot. The results showed that SHARPIN expression was increased in CCA tissue, and this was involved in cell proliferation. SHARPIN silencing resulted in accumulated reactive oxygen species, reduced mitochondrial fission, and a reduced mitochondrial membrane potential. Silencing of SHARPIN inhibited the ubiquitination and degradation of p53, and downregulated levels of SLC7A11, GPX4, SOD-1, and SOD-2, all of which contributed to excessive oxidative stress that leads to ferroptosis. Overexpression of SHARPIN would reverse the above process. The collected data suggest that in CCA, SHARPIN-mediated cell ferroptosis via the p53/SLC7A11/GPX4 signaling pathway is inhibited. Targeting SHARPIN might be a promising approach for the treatment of CCA.
Collapse
Affiliation(s)
- Chong Zeng
- Department of Medical Research Center, Shunde HospitalSouthern Medical University (The First People's Hospital of Shunde Foshan)FoshanChina
| | - Jie Lin
- Department of Hepatobiliary SurgeryShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanChina
| | - Ketao Zhang
- Department of Hepatobiliary SurgeryShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanChina
| | - Huohui Ou
- Department of Hepatobiliary SurgeryShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanChina
| | - Ke Shen
- Department of Pathology, Shunde HospitalSouthern Medical University (The First People's Hospital of Shunde)FoshanChina
| | - Qingbo Liu
- Department of Hepatobiliary SurgeryShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanChina
| | - Zibo Wei
- Department of Medical Research Center, Shunde HospitalSouthern Medical University (The First People's Hospital of Shunde Foshan)FoshanChina
| | - Xinhuai Dong
- Department of Medical Research Center, Shunde HospitalSouthern Medical University (The First People's Hospital of Shunde Foshan)FoshanChina
| | - Xiaokang Zeng
- Department of Medical Research Center, Shunde HospitalSouthern Medical University (The First People's Hospital of Shunde Foshan)FoshanChina
| | - Liming Zeng
- Department of Medical Research Center, Shunde HospitalSouthern Medical University (The First People's Hospital of Shunde Foshan)FoshanChina
| | - Weidong Wang
- Department of Hepatobiliary SurgeryShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanChina
| | - Jie Yao
- Department of Medical Research Center, Shunde HospitalSouthern Medical University (The First People's Hospital of Shunde Foshan)FoshanChina
| |
Collapse
|
29
|
Wu C, Shen Z, Lu Y, Sun F, Shi H. p53 Promotes Ferroptosis in Macrophages Treated with Fe 3O 4 Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2022; 14:42791-42803. [PMID: 36112832 DOI: 10.1021/acsami.2c00707] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Fe3O4 nanoparticles are the most widely used magnetic nanoparticles in the biomedicine field. The biodistribution of most nanoparticles in vivo is determined by the capture of macrophages; however, the effects of nanoparticles on macrophages remain poorly understood. Here, we demonstrated that Fe3O4 nanoparticles could reduce macrophage viability after 48 h of treatment and induce a shift in macrophage polarization toward the M1 phenotype; RNA sequencing revealed the activation of the ferroptosis pathway and p53 upregulation compared to the control group. The expression in p53, xCT, glutathione peroxidase 4 (GPX4), and transferrin receptor (TFR) in macrophages was similar to that in erastin-induced ferroptosis in macrophages, and the ultrastructural morphology of mitochondria was consistent with that of erastin-treated cells. We used DCFH-DA to estimate the intracellular reactive oxygen species content in Fe3O4 nanoparticles treated with Ana-1 and JC-1 fluorescent probes to detect the mitochondrial membrane potential change; both showed to be time-dependent. Fer-1 inhibited the reduction of the glutathione/oxidized glutathione (GSH/GSSG) ratio and inhibited intracellular oxidative stress states; therefore, Fe3O4 nanoparticles induced ferroptosis in macrophages. Finally, we used pifithrin-α hydrobromide (PFT) as a p53 inhibitor to verify whether the high expression of p53 is involved in mediating this process. After PFT treatment, the live/dead cell rate, TFR, p53 expression, and GPX4 consumption were inhibited and mitigated the GSH/GSSG ratio reduction as well. This indicates that p53 may contribute to Fe3O4 nanoparticle-induced ferroptosis of macrophages. We provide a theoretical basis for the molecular mechanisms of ferroptosis in macrophages and the biotoxicity in vivo induced by Fe3O4 nanoparticles.
Collapse
Affiliation(s)
- Cong Wu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| | - Zhiming Shen
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| | - Yi Lu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| | - Fei Sun
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| | - Hongcan Shi
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| |
Collapse
|
30
|
Xiao L, Huang H, Fan S, Zheng B, Wu J, Zhang J, Pi J, Xu JF. Ferroptosis: A mixed blessing for infectious diseases. Front Pharmacol 2022; 13:992734. [PMID: 36160441 PMCID: PMC9490125 DOI: 10.3389/fphar.2022.992734] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
To date, it has been confirmed that the occurrence and development of infectious diseases are tightly associated with regulatory cell death processes, such as apoptosis, autophagy, and necroptosis. Ferroptosis, as a newly discovered form of regulatory cell death characterized by iron-dependent lipid peroxidation, is not only closely associated with tumor progression, but is also found to be tightly related to the regulation of infectious diseases, such as Tuberculosis, Cryptococcal meningitis, Malaria and COVID-2019. The emerging critical roles of ferroptosis that has been found in infectious disease highlight ferroptosis as a potential therapeutic target in this field, which is therefore widely expected to be developed into new therapy strategy against infectious diseases. Here, we summarized the underlying mechanisms of ferroptosis and highlighted the intersections between host immunity and ferroptosis. Moreover, we illuminated the roles of ferroptosis in the occurrence and progression of different infectious diseases, which might provide some unique inspiration and thought-provoking perspectives for the future research of these infectious diseases, especially for the development of ferroptosis-based therapy strategy against infectious diseases.
Collapse
Affiliation(s)
- Leyao Xiao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Huanshao Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Shuhao Fan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Biying Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jianguo Wu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Junai Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
- *Correspondence: Jiang Pi, ; Jun-Fa Xu,
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
- *Correspondence: Jiang Pi, ; Jun-Fa Xu,
| |
Collapse
|
31
|
Yi L, Hu Y, Wu Z, Li Y, Kong M, Kang Z, Zuoyuan B, Yang Z. TFRC upregulation promotes ferroptosis in CVB3 infection via nucleus recruitment of Sp1. Cell Death Dis 2022; 13:592. [PMID: 35821227 PMCID: PMC9276735 DOI: 10.1038/s41419-022-05027-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 01/21/2023]
Abstract
CVB3 is a single positive-strand enterovirus, and a common pathogen in myocarditis etiology. Although a number of antiviral candidates are under development, specific targeted therapy is not available for CVB3. Ferroptosis is a new type of regulatory cell death discovered in recent years. In this study, our team provided the first evidence that ferroptosis existed in CVB3 infection in vivo and in vitro by iron overload, and massive accumulation of lipid peroxides. Mechanistically, we construct a classical model of HeLa cells following a time-course infection (6, 12, 24, 36, 48 h) with CVB3 (MOI = 10). We demonstrated that the TFRC gene plays an important role in promoting ferroptosis in CVB3 infection and downregulation of TFRC attenuated the ferroptosis. Interestingly, we observed that TFRC was nuclear translocation induced by the CVB3, which was predominantly localized in the cell membrane, but redistributed to the nucleus during CVB3 infection. Moreover, we found that the transcription factor Sp1 was an essential factor that could bind to the TFRC promoter and upregulate the TFRC transcription. Collectively, these results suggest that the Sp1/TFRC/Fe axis may provide a new target for the development of therapies against CVB3 infection.
Collapse
Affiliation(s)
- Lu Yi
- Department of Pediatrics, Third Xiangya Hospital of Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Yanan Hu
- Department of Pediatrics, Third Xiangya Hospital of Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Zhixiang Wu
- Department of Pediatrics, Third Xiangya Hospital of Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Ying Li
- Department of Pediatrics, Third Xiangya Hospital of Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Min Kong
- Department of Pediatrics, Third Xiangya Hospital of Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Zhijuan Kang
- Department of Pediatrics, Third Xiangya Hospital of Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Bojiao Zuoyuan
- Department of Pediatrics, Third Xiangya Hospital of Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Zuocheng Yang
- Department of Pediatrics, Third Xiangya Hospital of Central South University, 410013, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
32
|
Wagner MP, Formaglio P, Gorgette O, Dziekan JM, Huon C, Berneburg I, Rahlfs S, Barale JC, Feinstein SI, Fisher AB, Ménard D, Bozdech Z, Amino R, Touqui L, Chitnis CE. Human peroxiredoxin 6 is essential for malaria parasites and provides a host-based drug target. Cell Rep 2022; 39:110923. [PMID: 35705035 DOI: 10.1016/j.celrep.2022.110923] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/30/2022] [Accepted: 05/17/2022] [Indexed: 11/30/2022] Open
Abstract
The uptake and digestion of host hemoglobin by malaria parasites during blood-stage growth leads to significant oxidative damage of membrane lipids. Repair of lipid peroxidation damage is crucial for parasite survival. Here, we demonstrate that Plasmodium falciparum imports a host antioxidant enzyme, peroxiredoxin 6 (PRDX6), during hemoglobin uptake from the red blood cell cytosol. PRDX6 is a lipid-peroxidation repair enzyme with phospholipase A2 (PLA2) activity. Inhibition of PRDX6 with a PLA2 inhibitor, Darapladib, increases lipid-peroxidation damage in the parasite and disrupts transport of hemoglobin-containing vesicles to the food vacuole, causing parasite death. Furthermore, inhibition of PRDX6 synergistically reduces the survival of artemisinin-resistant parasites following co-treatment of parasite cultures with artemisinin and Darapladib. Thus, PRDX6 is a host-derived drug target for development of antimalarial drugs that could help overcome artemisinin resistance.
Collapse
Affiliation(s)
- Matthias Paulus Wagner
- Institut Pasteur, Université de Paris, Malaria Parasite Biology and Vaccines Unit, Paris, France
| | - Pauline Formaglio
- Institut Pasteur, Université de Paris, Malaria Infection and Immunity Unit, Paris, France
| | - Olivier Gorgette
- Institut Pasteur, Department of Cell Biology and Infection, Centre for Innovation and Technological Research, Ultrastructural Bioimaging Unit, Paris, France
| | - Jerzy Michal Dziekan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Christèle Huon
- Institut Pasteur, Université de Paris, Malaria Parasite Biology and Vaccines Unit, Paris, France
| | - Isabell Berneburg
- Biochemistry and Molecular Biology, Interdisciplinary Research Centre, Justus Liebig University Giessen, Giessen, Germany
| | - Stefan Rahlfs
- Biochemistry and Molecular Biology, Interdisciplinary Research Centre, Justus Liebig University Giessen, Giessen, Germany
| | - Jean-Christophe Barale
- Institut Pasteur, Université de Paris, CNRS UMR 3528, Structural Microbiology Unit, Paris, France; Institut Pasteur, Pasteur International Unit, Pasteur International Network, Malaria Translational Research Unit, Phnom Penh, Cambodia and Paris, France
| | | | - Aron B Fisher
- Peroxitech, Inc., Philadelphia, PA, USA; Institute for Environmental Medicine, Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Didier Ménard
- Institut Pasteur, Université de Paris, INSERM U1201, Malaria Genetics and Resistance Unit, Paris, France; Dynamics of Host-Pathogen Interactions, EA 7292, IPPTS, Strasbourg University, Strasbourg, France
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Rogerio Amino
- Institut Pasteur, Université de Paris, Malaria Infection and Immunity Unit, Paris, France
| | - Lhousseine Touqui
- Cystic Fibrosis, Physiopathology and Phenogenomics, INSERM Unit 938, Saint-Antoine, Paris, France; Institut Pasteur, Université de Paris, Laboratory of Cystic Fibrosis and Chronic Bronchopathies, Paris, France
| | - Chetan E Chitnis
- Institut Pasteur, Université de Paris, Malaria Parasite Biology and Vaccines Unit, Paris, France.
| |
Collapse
|
33
|
Zhou X, Fu Y, Liu W, Mu Y, Zhang H, Chen J, Liu P. Ferroptosis in Chronic Liver Diseases: Opportunities and Challenges. Front Mol Biosci 2022; 9:928321. [PMID: 35720113 PMCID: PMC9205467 DOI: 10.3389/fmolb.2022.928321] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/16/2022] [Indexed: 01/01/2023] Open
Abstract
Ferroptosis, an iron-dependent non-apoptotic cell death characterized by lipid peroxidation, is a cell death pathway discovered in recent years. Ferroptosis plays an important role in tumors, ischemia-reperfusion injury, neurological diseases, blood diseases, etc. Recent studies have shown the importance of ferroptosis in chronic liver disease. This article summarizes the pathological mechanisms of ferroptosis involved in System Xc-, iron metabolism, lipid metabolism, and some GPX4-independent pathways, and the latest research on ferroptosis in chronic liver diseases such as alcoholic liver disease, non-alcoholic fatty liver disease, liver fibrosis, hepatocellular carcinoma. In addition, the current bottleneck issues that restrict the research on ferroptosis are proposed to provide ideas and strategies for exploring new therapeutic targets for chronic liver diseases.
Collapse
Affiliation(s)
- Xiaoxi Zhou
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Yadong Fu
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Wei Liu
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Yongping Mu
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Hua Zhang
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Jiamei Chen
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Ping Liu
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
34
|
Hu M, Men Y, Chen L, Huang J, Duan F, Zhang Y, Dong S. Dexmedetomidine exerts its protective effect on cerebral ischemia reperfusion injury in mice by inhibiting ferroptosis. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:600-609. [PMID: 35753730 PMCID: PMC10929910 DOI: 10.11817/j.issn.1672-7347.2022.210443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVES Stroke is one of the major diseases that can threaten human life and health. The incidence of ischemic stroke accounts for more than 70% of stroke. The mechanism of ischemia reperfusion (IR) injury caused by ischemic stroke is extremely complex. In recent years, dexmedetomidine has been increasingly studied in anti-cerebral IR injury as a common clinical anesthetic adjunct, but its specific mechanism is not fully understood. Therefore, this study aims to explore the effects and mechanisms of dexmedetomidine on cerebral IR injury in mice. METHODS The mouse middle cerebral artery occlusion (MCAO) model was prepared by modified suture method. Male ICR mice were randomly divided into a sham group, an IR group, an IR+D1 group (IR+administered 25 µg/kg dexmedetomidine), an IR+D2 group(IR+administered 50 µg/kg dexmedetomidine), an IR+D3 group (IR+administered 100 µg/kg dexmedetomidine), and an IR+D2+ML385 group (IR+administered 50 µg/kg dexmedetomidine and 30 mg/kg ML385). The neurologic behavior of mice was evaluated by Longa's five-point method. 2,3,5-triphenyltetrazolium chloride (TTC) staining was used to detect the percentage of cerebral infarct volume in mice. The protein expressions of nuclear factor erythroid 2-related factor 2 (Nrf2), transferrin receptor 1 (TFR1), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11) in the cerebral tissues of mice were detected by Western blotting.Mitochondrial morphology was observed under the transmission electron microscope. The contents of MDA, Fe2+, and GSH in the cerebral tissues of mice were detected. RESULTS Compared with the sham group, neurobehavioral scores, cerebral infarct volume, the contents of MDA and Fe2+, as well as the protein expression of TFR1 were significantly increased; the contents of GSH and the protein expression of SLC7A11 and GPX4 were significantly reduced (all P<0.05); mitochondria in cerebral tissue were wrinkled, cristae were reduced, and membrane density was increased in the IR group. Compared with the IR group, neurobehavioral scores, cerebral infarction volume, MDA and Fe2+ contents, as well as the protein expression of TFR1 were significantly reduced; the contents of GSH and the protein expression of SLC7A11 and GPX4 were significantly increased (all P<0.05); mitochondrial damage in cerebral tissue was significantly relieved with the pre-treatment of dexmedetomidine. Compared with the IR+D2 group, neurobehavioral scores, cerebral infarction volume, MDA and Fe2+ contents, as well as the protein expression of TFR1 were significantly increased; the contents of GSH and the protein expression of SLC7A11 and GPX4 were significantly reduced (all P<0.05);mitochondria reappeared significantly damaged with the ML385 on the basis of dexmedetomidine pre-treatment. CONCLUSIONS The protective effect of dexmedetomidine on cerebral IR injury mice is related to its inhibition of ferroptosis, and the mechanism might be related to its regulation of Nrf2 expression.
Collapse
Affiliation(s)
- Miao Hu
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030.
| | - Yunzheng Men
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Lei Chen
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Fangfang Duan
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Yuxin Zhang
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Shuying Dong
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030.
- Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu Anhui 233030, China.
| |
Collapse
|
35
|
Matteucci KC, Correa AAS, Costa DL. Recent Advances in Host-Directed Therapies for Tuberculosis and Malaria. Front Cell Infect Microbiol 2022; 12:905278. [PMID: 35669122 PMCID: PMC9163498 DOI: 10.3389/fcimb.2022.905278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/21/2022] [Indexed: 11/30/2022] Open
Abstract
Tuberculosis (TB), caused by the bacterium Mycobacterium tuberculosis, and malaria, caused by parasites from the Plasmodium genus, are two of the major causes of death due to infectious diseases in the world. Both diseases are treatable with drugs that have microbicidal properties against each of the etiologic agents. However, problems related to treatment compliance by patients and emergence of drug resistant microorganisms have been a major problem for combating TB and malaria. This factor is further complicated by the absence of highly effective vaccines that can prevent the infection with either M. tuberculosis or Plasmodium. However, certain host biological processes have been found to play a role in the promotion of infection or in the pathogenesis of each disease. These processes can be targeted by host-directed therapies (HDTs), which can be administered in conjunction with the standard drug treatments for each pathogen, aiming to accelerate their elimination or to minimize detrimental side effects resulting from exacerbated inflammation. In this review we discuss potential new targets for the development of HDTs revealed by recent advances in the knowledge of host-pathogen interaction biology, and present an overview of strategies that have been tested in vivo, either in experimental models or in patients.
Collapse
Affiliation(s)
- Kely C. Matteucci
- Plataforma de Medicina Translacional Fundação Oswaldo Cruz/Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - André A. S. Correa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Diego L. Costa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Diego L. Costa,
| |
Collapse
|
36
|
Liu Y, Gu W. p53 in ferroptosis regulation: the new weapon for the old guardian. Cell Death Differ 2022; 29:895-910. [PMID: 35087226 PMCID: PMC9091200 DOI: 10.1038/s41418-022-00943-y] [Citation(s) in RCA: 318] [Impact Index Per Article: 106.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 02/08/2023] Open
Abstract
Although the conventional activities of p53 such as cell cycle arrest, senescence, and apoptosis are well accepted as the major checkpoints in stress responses, accumulating evidence implicates the importance of other tumor suppression mechanisms. Among these unconventional activities, an iron-dependent form of non-apoptotic cell death, termed ferroptosis, attracts great interest. Unlike apoptotic cell death, activation of p53 alone is not sufficient to induce ferroptosis directly; instead, through its metabolic targets, p53 is able to modulate the ferroptosis response in the presence of ferroptosis inducers such as GPX4 inhibitors or high levels of ROS. Here, we review the role of ferroptosis in p53-mediated tumor suppression, with a focus on what cellular factors are critical for p53-dependent ferroptosis during tumor suppression and how p53 modulates both the canonical (GPX4-dependent) and the non-canonical (GPX4-independent) ferroptosis pathways. We also discuss the possibility of targeting p53-mediated ferroptotic responses for the treatment of human cancers and potentially, other diseases.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
| |
Collapse
|
37
|
Plasmodium berghei-Mediated NRF2 Activation in Infected Hepatocytes Enhances Parasite Survival. Cell Microbiol 2022. [DOI: 10.1155/2022/7647976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The protozoan parasite Plasmodium, causative agent of malaria, initially invades and develops in hepatocytes where it resides in a parasitophorous vacuole (PV). A single invaded parasite develops into thousands of daughter parasites. Survival of the host cell is crucial for successful completion of liver stage development. Nuclear factor erythroid-derived 2-related factor 2 (NRF2) is a transcription factor known to induce transcription of cytoprotective genes when activated. Here we show that NRF2 is activated in Plasmodium berghei-infected hepatocytes. We observed that this NRF2 activation depends on PV membrane resident p62 recruiting KEAP1, the negative regulator of NRF2. Disrupting the NRF2 gene results in reduced parasite survival, indicating that NRF2 signaling is an important event for parasite development in hepatocytes. Together, our observations uncovered a novel mechanism of how Plasmodium parasites ensure host cell survival during liver stage development.
Collapse
|
38
|
Glennon EKK, Tongogara T, Primavera VI, Reeder SM, Wei L, Kaushansky A. Elucidating Spatially-Resolved Changes in Host Signaling During Plasmodium Liver-Stage Infection. Front Cell Infect Microbiol 2022; 11:804186. [PMID: 35111697 PMCID: PMC8801743 DOI: 10.3389/fcimb.2021.804186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022] Open
Abstract
Upon transmission to the human host, Plasmodium sporozoites exit the skin, are taken up by the blood stream, and then travel to the liver where they infect and significantly modify a single hepatocyte. Low infection rates within the liver have made proteomic studies of infected hepatocytes challenging, particularly in vivo, and existing studies have been largely unable to consider how protein and phosphoprotein differences are altered at different spatial locations within the heterogeneous liver. Using digital spatial profiling, we characterized changes in host signaling during Plasmodium yoelii infection in vivo without disrupting the liver tissue. Moreover, we measured alterations in protein expression around infected hepatocytes and identified a subset of CD163+ Kupffer cells that migrate towards infected cells during infection. These data offer the first insight into the heterogeneous microenvironment that surrounds the infected hepatocyte and provide insights into how the parasite may alter its milieu to influence its survival and modulate immunity.
Collapse
Affiliation(s)
- Elizabeth K. K. Glennon
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Tinotenda Tongogara
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Grinnell College, Grinnell, IA, United States
| | - Veronica I. Primavera
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Sophia M. Reeder
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Ling Wei
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Alexis Kaushansky
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Brotman Baty Institute for Precision Medicine, Seattle, WA, United States
- *Correspondence: Alexis Kaushansky,
| |
Collapse
|
39
|
Zhang S, Xin W, Anderson GJ, Li R, Gao L, Chen S, Zhao J, Liu S. Double-edge sword roles of iron in driving energy production versus instigating ferroptosis. Cell Death Dis 2022; 13:40. [PMID: 35013137 PMCID: PMC8748693 DOI: 10.1038/s41419-021-04490-1] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022]
Abstract
Iron is vital for many physiological functions, including energy production, and dysregulated iron homeostasis underlies a number of pathologies. Ferroptosis is a recently recognized form of regulated cell death that is characterized by iron dependency and lipid peroxidation, and this process has been reported to be involved in multiple diseases. The mechanisms underlying ferroptosis are complex, and involve both well-described pathways (including the iron-induced Fenton reaction, impaired antioxidant capacity, and mitochondrial dysfunction) and novel interactions linked to cellular energy production. In this review, we examine the contribution of iron to diverse metabolic activities and their relationship to ferroptosis. There is an emphasis on the role of iron in driving energy production and its link to ferroptosis under both physiological and pathological conditions. In conclusion, excess reactive oxygen species production driven by disordered iron metabolism, which induces Fenton reaction and/or impairs mitochondrial function and energy metabolism, is a key inducer of ferroptosis.
Collapse
Affiliation(s)
- Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Wei Xin
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Gregory J Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Ruibin Li
- School for Radiological and Interdisciplinary Science, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, 250031, China
| | - Shuguang Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, 250031, China.
| | - Sijin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China.
| |
Collapse
|
40
|
MiR-34c-3p upregulates erastin-induced ferroptosis to inhibit proliferation in oral squamous cell carcinomas by targeting SLC7A11. Pathol Res Pract 2022; 231:153778. [DOI: 10.1016/j.prp.2022.153778] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 01/20/2022] [Accepted: 01/22/2022] [Indexed: 12/14/2022]
|
41
|
Zhang W, Liu J, Li X, Zheng Y, Chen L, Wang D, Foda MF, Ma Z, Zhao Y, Han H. Precise Chemodynamic Therapy of Cancer by Trifunctional Bacterium-Based Nanozymes. ACS NANO 2021; 15:19321-19333. [PMID: 34851608 DOI: 10.1021/acsnano.1c05605] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Chemodynamic therapy (CDT) destroys cancer cells by converting H2O2 or O2 into reactive oxygen species (ROS), but its therapeutic efficacy is restricted by the antioxidant capacity of tumor. Previous solutions focused on strengthening the nanodrugs with the ability to increase ROS production or weaken the antioxidant capacity of cancer cells. Conversely, we here develop a mild nanodrug with negligible side effects. Specifically, the Au@Pt nanozyme decorated on a bacterial surface (Bac-Au@Pt) is reported to achieve precise CDT. Due to the tumor targeting ability of bacteria and catalytic property of Au@Pt nanozyme under acidic conditions, this nanosystem can release ROS to tumor cells effectively. In addition, the interferon gamma released by T cells specifically decreases the intracellular reductants in tumor cells, while having no obvious effect on normal cells. Therefore, a low dose of Bac-Au@Pt achieves a satisfactory therapeutic efficacy to tumor cells and is nontoxic to normal cells even at their acidic components. This nanosystem enables CDT and immunotherapy to mutually benefit and improve by each other, providing a promising strategy to achieve high anticancer efficacy even with a low dose usage.
Collapse
Affiliation(s)
- Weiyun Zhang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
- College of Science, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jiawei Liu
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Xuyu Li
- College of Science, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yue Zheng
- Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Lianfu Chen
- South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510520, PR China
| | - Dongdong Wang
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Mohamed Frahat Foda
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
- Department of Biochemistry, Faculty of Agriculture, Benha University, Moshtohor, Toukh 13736, Egypt
| | - Zhaoyu Ma
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Heyou Han
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
- College of Science, Huazhong Agricultural University, Wuhan 430070, PR China
| |
Collapse
|
42
|
Ferroptosis in Non-Small Cell Lung Cancer: Progression and Therapeutic Potential on It. Int J Mol Sci 2021; 22:ijms222413335. [PMID: 34948133 PMCID: PMC8704137 DOI: 10.3390/ijms222413335] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/28/2021] [Accepted: 12/09/2021] [Indexed: 12/16/2022] Open
Abstract
As a main subtype of lung cancer, the current situation of non-small cell lung cancer (NSCLC) remains severe worldwide with a 19% survival rate at 5 years. As the conventional therapy approaches, such as chemotherapy, radiotherapy, targeted therapy, and immunotherapy, gradually develop into therapy resistance, searching for a novel therapeutic strategy for NSCLC is urgent. Ferroptosis, an iron-dependent programmed necrosis, has now been widely considered as a key factor affecting the tumorigenesis and progression in various cancers. Focusing on its effect in NSCLC, in different situations, ferroptosis can be triggered or restrained. When ferroptosis was induced in NSCLC, it was available to inhibit the tumor progression both in vitro and in vivo. The dominating mechanism was due to a regulation of the classic ferroptosis-repressed GSH-dependent GPX4 signaling pathway instead of other fractional regulating signal axes that regulated ferroptosis via impacting on the ROS, cellular iron levels, etc. In terms of the prevention of ferroptosis in NSCLC, an GSH-independent mechanism was also discovered, interestingly exhibiting the same upstream as the GPX4 signaling. In addition, this review summarizes the progression of ferroptosis in NSCLC and elaborates their association and specific mechanisms through bioinformatics analysis with multiple experimental evidence from different cascades. Finally, this review also points out the possibility of ferroptosis working as a novel strategy for therapy resistance in NSCLC, emphasizing its therapeutic potential.
Collapse
|
43
|
Olanlokun JO, Abiodun WO, Ebenezer O, Koorbanally NA, Olorunsogo OO. Curcumin modulates multiple cell death, matrix metalloproteinase activation and cardiac protein release in susceptible and resistant Plasmodium berghei-infected mice. Biomed Pharmacother 2021; 146:112454. [PMID: 34894518 DOI: 10.1016/j.biopha.2021.112454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 01/08/2023] Open
Abstract
Pro-inflammatory signaling, cell death, and metalloproteinases activation are events in Plasmodium infection. However, it is not known if treatment with mefloquine (MF), and curcumin (CM) supplementation, will modulate these conditions. Malaria was induced in two different studies using susceptible (NK 65, study 1) and resistant (ANKA, study 2) strains of mouse malaria parasites (Plasmodium berghei) in thirty male Swiss mice (n = 5) in each study. Following confirmation of parasitemia, mice received 10 mL/kg distilled water (infected control), MF (10 mg/kg), MF and CM (25 mg/kg), MF and CM (50 mg/kg), CM (25 mg/kg) and CM (50 mg/kg). Five mice (not infected) were used as control. After treatment, the animals were sacrificed, serum obtained and liver mitochondria were isolated. Serum Tumour Necrosis Factor alpha (TNF-α), C-reactive protein (CRP), Interleukins-1 beta (IL-1β) and Interleukins-6 (IL-6) as well as caspases-3, 9 (C3 and C9), p53, serum troponin I (TI) and creatine kinase (CK), were assayed using ELISA techniques. Mitochondrial membrane permeability transition (mPT) pore opening, mitochondrial F0F1 ATPase activity, and lipid peroxidation (mLPO) were determined spectrophotometrically. Matrix metalloproteinases 2 (MMP-2) and 9 (MMP-9) expressions were determined using electrophoresis. CM supplementation (25 mg/kg) significantly decreased serum p53, TNF-α, CRP and IL-6 compared with MF. In the resistant model, CM prevented mPT pore opening, significantly decreased F0F1 ATPase activity and mLPO. MF activated caspase-3 while supplementation with CM significantly decreased this effect. Furthermore, MMP-2 and MMP-9 were selectively expressed in the susceptible model. Malarial treatment with mefloquine elicits different cell death responses while supplementation with curcumin decreased TI level and CK activities.
Collapse
Affiliation(s)
- John O Olanlokun
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Nigeria.
| | - Wisdom Oshireku Abiodun
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Nigeria
| | - Oluwakemi Ebenezer
- Faculty of Natural Science, Department of Chemistry, Mangosuthu University of Technology, 511 Mangosuthu Highway, Durban 4000, South Africa
| | - Neil A Koorbanally
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Olufunso Olabode Olorunsogo
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Nigeria
| |
Collapse
|
44
|
Zhang L, Wang F, Li D, Yan Y, Wang H. Transferrin receptor-mediated reactive oxygen species promotes ferroptosis of KGN cells via regulating NADPH oxidase 1/PTEN induced kinase 1/acyl-CoA synthetase long chain family member 4 signaling. Bioengineered 2021; 12:4983-4994. [PMID: 34369274 PMCID: PMC8806504 DOI: 10.1080/21655979.2021.1956403] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/13/2021] [Indexed: 12/20/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age. Abnormal ovarian folliculogenesis is the main factor responsible for PCOS. Iron metabolism plays a vital role in endocrine disorder. This study aimed to investigate the potentials of iron metabolism in PCOS and the underlying molecular mechanisms. Mice were injected with dehydroepiandrosterone (DHEA) to establish the PCOS model in-vivo. H & E staining was performed for histological analysis; qRT-PCR and western blot were employed to determine the mRNA and protein expressions. Immunofluorescence was used for mitochondrial staining. Cellular functions were detected using CCK-8 and PI staining assays. Ferric ammonium citrate (FAC) activates the transferrin receptor (TFRC), increases the iron content, and suppresses the cell viability of the human granulosa-like tumor cell line (KGN). However, TFRC knockdown suppressed ferroptosis of KGN cells. Iron uptake mediated the activation of NADPH oxidase 1 (NOX1) signaling, which induced the release of reactive oxygen species (ROS) and mitochondrial damage. Moreover, TFRC activated PTEN induced kinase 1 (PINK1) signaling and induced mitophagy; iron-uptake-induced upregulation of acyl-CoA synthetase long chain family member 4 (ACSL4) was required for mitophagy activation and glutathione peroxidase 4 (GPX4) degradation. Additionally, FAC increased iron uptake and suppressed the folliculogenesis in-vivo. In conclusion, TFRC increased the iron content, mediated the release of ROS, activated mitophagy, and induced lipid peroxidation, which further promoted the ferroptosis of KGN cells. Therefore, the inhibitory effects of TFRC/NOX1/PINK1/ACSL4 signaling on folliculogenesis can be a potential target for PCOS.[Figure: see text].
Collapse
Affiliation(s)
- Lingzhi Zhang
- Department of Obstetrics and Gynecology, Cao County People’s Hospital, Shandong, China
| | - Fang Wang
- Department of Obstetrics and Gynecology, Cao County People’s Hospital, Shandong, China
| | - Dongmei Li
- Department of Obstetrics and Gynecology, Cao County People’s Hospital, Shandong, China
| | - Yufeng Yan
- Department of Obstetrics and Gynecology, Cao County People’s Hospital, Shandong, China
| | - Hongyan Wang
- Department of Obstetrics and Gynecology, Cao County People’s Hospital, Shandong, China
| |
Collapse
|
45
|
Abstract
Host-directed therapy (HDT) is gaining traction as a strategy to combat infectious diseases caused by viruses and intracellular bacteria, but its implementation in the context of parasitic diseases has received less attention. Here, we provide a brief overview of this field and advocate HDT as a promising strategy for antimalarial intervention based on untapped targets. HDT provides a basis from which repurposed drugs could be rapidly deployed and is likely to strongly limit the emergence of resistance. This strategy can be applied to any intracellular pathogen and is particularly well placed in situations in which rapid identification of treatments is needed, such as emerging infections and pandemics, as starkly illustrated by the current COVID-19 crisis.
Collapse
|
46
|
Emerging role of ferroptosis in breast cancer: New dawn for overcoming tumor progression. Pharmacol Ther 2021; 232:107992. [PMID: 34606782 DOI: 10.1016/j.pharmthera.2021.107992] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer has become a serious threat to women's health. Cancer progression is mainly derived from resistance to apoptosis induced by procedures or therapies. Therefore, new drugs or models that can overcome apoptosis resistance should be identified. Ferroptosis is a recently identified mode of cell death characterized by excess reactive oxygen species-induced lipid peroxidation. Since ferroptosis is distinct from apoptosis, necrosis and autophagy, its induction successfully eliminates cancer cells that are resistant to other modes of cell death. Therefore, ferroptosis may become a new direction around which to design breast cancer treatment. Unfortunately, the complete appearance of ferroptosis in breast cancer has not yet been fully elucidated. Furthermore, whether ferroptosis inducers can be used in combination with traditional anti- breast cancer drugs is still unknown. Moreover, a summary of ferroptosis in breast cancer progression and therapy is currently not available. In this review, we discuss the roles of ferroptosis-associated modulators glutathione, glutathione peroxidase 4, iron, nuclear factor erythroid-2 related factor-2, superoxide dismutases, lipoxygenase and coenzyme Q in breast cancer. Furthermore, we provide evidence that traditional drugs against breast cancer induce ferroptosis, and that ferroptosis inducers eliminate breast cancer cells. Finally, we put forward prospect of using ferroptosis inducers in breast cancer therapy, and predict possible obstacles and corresponding solutions. This review will deepen our understanding of the relationship between ferroptosis and breast cancer, and provide new insights into breast cancer-related therapeutic strategies.
Collapse
|
47
|
The role of iron homeostasis in remodeling immune function and regulating inflammatory disease. Sci Bull (Beijing) 2021; 66:1806-1816. [PMID: 36654387 DOI: 10.1016/j.scib.2021.02.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/31/2020] [Accepted: 01/28/2021] [Indexed: 02/03/2023]
Abstract
The essential trace element iron regulates a wide range of biological processes in virtually all living organisms. Because both iron deficiency and iron overload can lead to various pathological conditions, iron homeostasis is tightly regulated, and understanding this complex process will help pave the way to developing new therapeutic strategies for inflammatory disease. In recent years, significant progress has been made with respect to elucidating the roles of iron and iron-related genes in the development and maintenance of the immune system. Here, we review the timing and mechanisms by which systemic and cellular iron metabolism are regulated during the inflammatory response and during infectious disease, processes in which both the host and the pathogen compete for iron. We also discuss the evidence and implications that immune cells such as macrophages, T cells, and B cells require sufficient amounts of iron for their proliferation and for mediating their effector functions, in which iron serves as a co-factor in toll-like receptor 4 (TLR4) signaling, mitochondrial respiration, posttranslational regulation, and epigenetic modification. In addition, we discuss the therapeutic implications of targeting ferroptosis, iron homeostasis and/or iron metabolism with respect to conferring protection against pathogen infection, controlling inflammation, and improving the efficacy of immunotherapy.
Collapse
|
48
|
Bagayoko S, Leon-Icaza SA, Pinilla M, Hessel A, Santoni K, Péricat D, Bordignon PJ, Moreau F, Eren E, Boyancé A, Naser E, Lefèvre L, Berrone C, Iakobachvili N, Metais A, Rombouts Y, Lugo-Villarino G, Coste A, Attrée I, Frank DW, Clevers H, Peters PJ, Cougoule C, Planès R, Meunier E. Host phospholipid peroxidation fuels ExoU-dependent cell necrosis and supports Pseudomonas aeruginosa-driven pathology. PLoS Pathog 2021; 17:e1009927. [PMID: 34516571 PMCID: PMC8460005 DOI: 10.1371/journal.ppat.1009927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 09/23/2021] [Accepted: 08/29/2021] [Indexed: 11/20/2022] Open
Abstract
Regulated cell necrosis supports immune and anti-infectious strategies of the body; however, dysregulation of these processes drives pathological organ damage. Pseudomonas aeruginosa expresses a phospholipase, ExoU that triggers pathological host cell necrosis through a poorly characterized pathway. Here, we investigated the molecular and cellular mechanisms of ExoU-mediated necrosis. We show that cellular peroxidised phospholipids enhance ExoU phospholipase activity, which drives necrosis of immune and non-immune cells. Conversely, both the endogenous lipid peroxidation regulator GPX4 and the pharmacological inhibition of lipid peroxidation delay ExoU-dependent cell necrosis and improve bacterial elimination in vitro and in vivo. Our findings also pertain to the ExoU-related phospholipase from the bacterial pathogen Burkholderia thailandensis, suggesting that exploitation of peroxidised phospholipids might be a conserved virulence mechanism among various microbial phospholipases. Overall, our results identify an original lipid peroxidation-based virulence mechanism as a strong contributor of microbial phospholipase-driven pathology.
Collapse
Affiliation(s)
- Salimata Bagayoko
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Stephen Adonai Leon-Icaza
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Miriam Pinilla
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Audrey Hessel
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Karin Santoni
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - David Péricat
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Pierre-Jean Bordignon
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Flavie Moreau
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
- Level 3 Biosafety Animal Core facility, Anexplo platform, Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Elif Eren
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Aurélien Boyancé
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Emmanuelle Naser
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
- Cytometry & Imaging Core facility, Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Lise Lefèvre
- RESTORE institute, University of Toulouse, CNRS, Toulouse, France
| | - Céline Berrone
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
- Level 3 Biosafety Animal Core facility, Anexplo platform, Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Nino Iakobachvili
- Division of Nanoscopy, Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | - Arnaud Metais
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Yoann Rombouts
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Geanncarlo Lugo-Villarino
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Agnès Coste
- RESTORE institute, University of Toulouse, CNRS, Toulouse, France
| | - Ina Attrée
- Univ. Grenoble Alpes, CNRS, CEA, IBS, Bacterial Pathogenesis and Cellular Responses, Grenoble, France
| | - Dara W. Frank
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, Netherlands
| | - Peter J. Peters
- Division of Nanoscopy, Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | - Céline Cougoule
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Rémi Planès
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| |
Collapse
|
49
|
Skorokhod O, Barrera V, Mandili G, Costanza F, Valente E, Ulliers D, Schwarzer E. Malaria Pigment Hemozoin Impairs GM-CSF Receptor Expression and Function by 4-Hydroxynonenal. Antioxidants (Basel) 2021; 10:antiox10081259. [PMID: 34439507 PMCID: PMC8389202 DOI: 10.3390/antiox10081259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/30/2021] [Accepted: 08/04/2021] [Indexed: 11/16/2022] Open
Abstract
Malarial pigment hemozoin (HZ) generates the lipoperoxidation product 4-hydroxynonenal (4-HNE), which is known to cause dysregulation of the immune response in malaria. The inhibition of granulocyte macrophage colony-stimulating factor (GM-CSF)-dependent differentiation of dendritic cells (DC) by HZ and 4-HNE was previously described in vitro, and the GM-CSF receptor (GM-CSF R) was hypothesised to be a primary target of 4-HNE in monocytes. In this study, we show the functional impact of HZ on GM-CSF R in monocytes and monocyte-derived DC by (i) impairing GM-CSF binding by 50 ± 9% and 65 ± 14%, respectively (n = 3 for both cell types); (ii) decreasing the expression of GM-CSF R functional subunit (CD116) on monocyte’s surface by 36 ± 11% (n = 6) and in cell lysate by 58 ± 16% (n = 3); and (iii) binding of 4-HNE to distinct amino acid residues on CD116. The data suggest that defective DC differentiation in malaria is caused by GM-CSF R dysregulation and GM-CSF R modification by lipoperoxidation product 4-HNE via direct interaction with its CD116 subunit.
Collapse
Affiliation(s)
- Oleksii Skorokhod
- Department of Life Sciences and Systems Biology, University of Torino, 10123 Torino, Italy
- Correspondence:
| | - Valentina Barrera
- Department of Oncology, University of Torino, 10126 Torino, Italy; (V.B.); (G.M.); (F.C.); (E.V.); (D.U.); (E.S.)
- National Health System Blood and Transplant, 14 Estuary Banks, Liverpool GB-L24 8RB, UK
| | - Giorgia Mandili
- Department of Oncology, University of Torino, 10126 Torino, Italy; (V.B.); (G.M.); (F.C.); (E.V.); (D.U.); (E.S.)
| | - Federica Costanza
- Department of Oncology, University of Torino, 10126 Torino, Italy; (V.B.); (G.M.); (F.C.); (E.V.); (D.U.); (E.S.)
| | - Elena Valente
- Department of Oncology, University of Torino, 10126 Torino, Italy; (V.B.); (G.M.); (F.C.); (E.V.); (D.U.); (E.S.)
| | - Daniela Ulliers
- Department of Oncology, University of Torino, 10126 Torino, Italy; (V.B.); (G.M.); (F.C.); (E.V.); (D.U.); (E.S.)
| | - Evelin Schwarzer
- Department of Oncology, University of Torino, 10126 Torino, Italy; (V.B.); (G.M.); (F.C.); (E.V.); (D.U.); (E.S.)
| |
Collapse
|
50
|
Lücke J, Sabihi M, Zhang T, Bauditz LF, Shiri AM, Giannou AD, Huber S. The good and the bad about separation anxiety: roles of IL-22 and IL-22BP in liver pathologies. Semin Immunopathol 2021; 43:591-607. [PMID: 33851257 PMCID: PMC8443499 DOI: 10.1007/s00281-021-00854-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022]
Abstract
The human liver fulfills several vital tasks daily and possesses an impressive ability to self-regenerate. However, the capacity of this self-healing process can be exhausted by a variety of different liver diseases, such as alcoholic liver damage, viral hepatitis, or hepatocellular carcinoma. Over time, all these diseases generally lead to progressive liver failure that can become fatal if left untreated. Thus, a great effort has been directed towards the development of innovative therapies. The most recently discovered therapies often involve modifying the patient's immune system to enhance a beneficial immune response. Current data suggest that, among others, the cytokine IL-22 might be a promising therapeutical candidate. IL-22 and its endogenous antagonist, IL-22BP, have been under thorough scientific investigation for nearly 20 years. While IL-22 is mainly produced by TH22 cells, ILC3s, NKT cells, or γδ T cells, sources of IL-22BP include dendritic cells, eosinophils, and CD4+ cells. In many settings, IL-22 was shown to promote regenerative potential and, thus, could protect tissues from pathogens and damage. However, the effects of IL-22 during carcinogenesis are more ambiguous and depend on the tumor entity and microenvironment. In line with its capabilities of neutralizing IL-22 in vivo, IL-22BP possesses often, but not always, an inverse expression pattern compared to its ligand. In this comprehensive review, we will summarize past and current findings regarding the roles of IL-22 and IL-22BP in liver diseases with a particular focus on the leading causes of advanced liver failure, namely, liver infections, liver damage, and liver malignancies.
Collapse
Affiliation(s)
- Jöran Lücke
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Morsal Sabihi
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Tao Zhang
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Lennart Fynn Bauditz
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ahmad Mustafa Shiri
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Anastasios D Giannou
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|