1
|
Wu C, Xu Z, Wang Q, Guo H, He X, Lin Y, Li L, Feng L, Li Q, Tang C. Investigation of Ferroptosis Mechanisms in Ischemic Stroke Treated with Electroacupuncture: Focusing on the NCOA4-FTH1 Signaling Pathway. Neurochem Res 2025; 50:137. [PMID: 40189641 DOI: 10.1007/s11064-025-04390-2] [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: 01/04/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025]
Abstract
Ischemic stroke remains a primary cause of mortality and morbidity, with ferroptosis emerging as a critical mechanism underlying neuronal damage post-ischemic injury. This study aims to elucidate the mechanisms of ferroptosis in ischemic stroke and assess the therapeutic potential of electroacupuncture, with emphasis on the NCOA4-FTH1 signaling pathway. After establishing a mouse model of middle cerebral artery occlusion (MCAO), we employed a combination of behavioral assessments and molecular techniques, including transmission electron microscopy, immunofluorescence, and Western blotting, to investigate the impact of electroacupuncture on ferroptosis markers. In addition, we constructed in vivo models of NCOA4 gene silencing and overexpression using adeno-associated virus (AAV) to verify whether electroacupuncture modulates the mechanism of ischemic stroke ferroptosis via the NCOA4-FTH1 signaling pathway. Our findings indicated that electroacupuncture could significantly downregulate NCOA4 expression while upregulating FTH1 and GPX4 levels in affected brain regions of MCAO mice. This resulted in reduced MDA levels, decreased iron ion concentration, a smaller brain infarct area, and improved motor function (p < 0.05). After constructing in vivo models of AAV-mediated NCOA4 gene silencing and overexpression, we demonstrated that electroacupuncture could attenuate iron deposition and inhibit ferroptosis in neurons by suppressing NCOA4 and upregulating FTH1, thereby ameliorating neurological deficits in the ischemic stroke model. These results suggest that electroacupuncture modulates ferroptosis through the NCOA4-FTH1 pathway, offering a novel therapeutic approach for neuroprotection following ischemic stroke.
Collapse
Affiliation(s)
- Chunxiao Wu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518104, P. R. China.
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, Guangdong Province, P. R. China.
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine; Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, P. R. China.
| | - Zhirui Xu
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 510130, Guangdong Province, P. R. China
| | - Qizhang Wang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518104, P. R. China
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, Guangdong Province, P. R. China
| | - Hongji Guo
- School of Medicine and Health, Shunde Polytechnic, Foshan, 528000, China
| | - Xin He
- Guangzhou Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, Guangdong Province, P. R. China
| | - Yuexi Lin
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, P. R. China
| | - Luping Li
- Guangzhou City Construction College, Guangzhou, Guangdong Province, P. R. China
| | - Linling Feng
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, P. R. China
| | - Qingyou Li
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, P. R. China
| | - Chunzhi Tang
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, P. R. China
| |
Collapse
|
2
|
Yuan D, Chen S, Zhou Y, Cao J, Zhang H, Wang Y, Zhou J. Ferritin1-mediated ferroptosis participates in granular acini degeneration of Haemaphysalis longicornis salivary glands. Ticks Tick Borne Dis 2025; 16:102464. [PMID: 40064061 DOI: 10.1016/j.ttbdis.2025.102464] [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: 07/17/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
Ticks (Haemaphysalis longicornis) transmit pathogens to their hosts through their salivary glands during blood-feeding. The salivary glands of adult parthenogenetic H. longicornis undergo degeneration post-engorgement. Clarifying the molecular mechanisms underlying salivary gland degeneration of H. longicornis is conducive to identifying novel targets for preventing and controlling these widespread vectors. In this study, we investigated the salivary glands of adult parthenogenetic H. longicornis to elucidate the relationship between ferroptosis, iron-dependent cell death, H. longicornis ferritin 1 (HlFer1) and salivary gland degeneration post-attachment and post-engorgement. Fluorescence microscopy, revealed increased iron accumulation, reactive oxygen species, lipid peroxidation, and decreased mitochondrial cristae in the granular acini of H. longicornis salivary glands post-engorgement. The results of a qPCR analysis indicated that HlFer1, glutathione peroxidase 4 (GPX4), transferrin (TRF), and high mobility group protein B1 (HMGB1) expression elevated in H. longicornis salivary glands post-attachment and post-engorgement. In vitro culture of H. longicornis salivary glands showed that erastin promotes ferroptosis, while ferrostatin-1 blocks this process. RNA interference (RNAi) targeting HlFer1 promoted ferroptosis in salivary gland granular acini. In conclusion, we demonstrated that HlFer1-induced ferroptosis is a key molecular mechanism underlying the salivary gland granular acini degeneration of H. longicornis. Our findings are important for developing novel preventive measures against H. longicornis as a disease vector.
Collapse
Affiliation(s)
- Dongqi Yuan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Songqin Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yongzhi Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jie Cao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Houshuang Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yanan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| | - Jinlin Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| |
Collapse
|
3
|
Rostami Ravari N, Sadri F, Mahdiabadi MA, Mohammadi Y, Ourang Z, Rezaei Z. Ferroptosis and noncoding RNAs: exploring mechanisms in lung cancer treatment. Front Cell Dev Biol 2025; 13:1522873. [PMID: 40078365 PMCID: PMC11897296 DOI: 10.3389/fcell.2025.1522873] [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: 11/05/2024] [Accepted: 01/31/2025] [Indexed: 03/14/2025] Open
Abstract
Lung cancer (LC) is a highly prevalent and deadly type of cancer characterized by intricate molecular pathways that drive tumor development, metastasis, and resistance to conventional treatments. Recently, ferroptosis, a controlled mechanism of cell death instigated by iron-dependent lipid peroxidation, has gained attention for its role in LC progression and treatment. Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are emerging as key modulators of ferroptosis, significantly influencing LC biology. This review explores how ncRNAs control ferroptotic pathways and affect tumor growth, metastasis, and therapy resistance in LC. By understanding the dual functions of ncRNAs in both activating and inhibiting ferroptosis, we aim to uncover new therapeutic targets and strategies for LC. These insights provide a promising direction for the development of ncRNA-based treatments designed to induce ferroptosis, potentially improving therapeutic outcomes for patients with LC.
Collapse
Affiliation(s)
- Nadi Rostami Ravari
- Department of Animal Science Researches, Agriculture and Natural Resources Education and Research Center of Kerman, Agriculture and Natural Resources Education and Research Organization (AREEO), Kerman, Iran
| | - Farzad Sadri
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Geriatric Health Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Ali Mahdiabadi
- Department of Internal Medicine, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Yaser Mohammadi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Ourang
- Department of Biochemistry, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Zohreh Rezaei
- Geriatric Health Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran
| |
Collapse
|
4
|
Zhao Y, Yang M, Liang X. The role of mitochondria in iron overload-induced damage. J Transl Med 2024; 22:1057. [PMID: 39587666 PMCID: PMC11587765 DOI: 10.1186/s12967-024-05740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/06/2024] [Indexed: 11/27/2024] Open
Abstract
Iron overload is a pathological condition characterized by the abnormal accumulation of iron within the body, which may result from excessive iron intake, disorders of iron metabolism, or specific disease states. This condition can lead to significant health complications and may pose life-threatening risks. The excessive accumulation of iron can induce cellular stress, adversely affecting the structure and function of mitochondria, thereby compromising overall organ function. Given the critical role of mitochondria in cellular metabolism and homeostasis, it is imperative to investigate how mitochondrial dysfunction induced by iron overload contributes to disease progression, as well as to explore mitochondrial-related pathways as potential therapeutic targets for various iron overload disorders. This review examines the mechanisms by which mitochondria are implicated in iron overload-induced damage, including increased oxidative stress, mitochondrial DNA damage, and disruptions in energy metabolism. Additionally, it addresses the relationship between these processes and various forms of programmed cell death, as well as alterations in mitochondrial dynamics. Furthermore, the review discusses strategies aimed at alleviating and mitigating the complications associated with iron overload in patients by targeting mitochondrial pathways.
Collapse
Affiliation(s)
- Yangyang Zhao
- Department of Transfusion, Affiliated Hospital of North Sichuan Medical college, Nanchong, Sichuan, P.R. China
| | - Mengjiao Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical college, Nanchong, Sichuan, P.R. China
- Graduate School of Comprehensive Human Science, University of Tsukuba, Tsukuba, Japan
| | - Xiaoxue Liang
- Chengdu Qingbaijiang District People's Hospital, Chengdu, 610300, Sichuan, P.R. China.
| |
Collapse
|
5
|
Tian L, Liu Q, Guo H, Zang H, Li Y. Fighting ischemia-reperfusion injury: Focusing on mitochondria-derived ferroptosis. Mitochondrion 2024; 79:101974. [PMID: 39461581 DOI: 10.1016/j.mito.2024.101974] [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: 07/04/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of mortality and morbidity. Current treatments for IRI have limited efficacy and novel therapeutic strategies are needed. Mitochondrial dysfunction not only initiates IRI but also plays a significant role in ferroptosis pathogenesis. Recent studies have highlighted that targeting mitochondrial pathways is a promising therapeutic approach for ferroptosis-induced IRI. The association between ferroptosis and IRI has been reviewed many times, but our review provides the first comprehensive overview with a focus on recent mitochondrial research. First, we present the role of mitochondria in ferroptosis. Then, we summarize the evidence on mitochondrial manipulation of ferroptosis in IRI and review recent therapeutic strategies aimed at targeting mitochondria-related ferroptosis to mitigate IRI. We hope our review will provide new ideas for the treatment of IRI and accelerate the transition from bench to bedside.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, China
| | - Hong Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Honggang Zang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
6
|
Huang X, Wang X, Wang Y, Shen S, Chen W, Liu T, Wang P, Fan X, Liu L, Jia J, Cong M. TIMP-1 Promotes Expression of MCP-1 and Macrophage Migration by Inducing Fli-1 in Experimental Liver Fibrosis. J Clin Transl Hepatol 2024; 12:634-645. [PMID: 38993513 PMCID: PMC11233975 DOI: 10.14218/jcth.2023.00514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 07/13/2024] Open
Abstract
Background and Aims Tissue inhibitor of metalloproteinase-1 (TIMP-1) plays a role in the excessive generation of extracellular matrix in liver fibrosis. This study aimed to explore the pathways through which TIMP-1 controls monocyte chemoattractant protein-1 (MCP-1) expression and promotes hepatic macrophage recruitment. Methods Liver fibrosis was triggered through carbon tetrachloride, and an adeno-associated virus containing small interfering RNA targeting TIMP-1 (siRNA-TIMP-1) was administered to both rats and mice. We assessed the extent of fibrosis and macrophage recruitment. The molecular mechanisms regulating macrophage recruitment by TIMP-1 were investigated through transwell migration assays, luciferase reporter assays, the use of pharmacological modulators, and an analysis of extracellular vesicles (EVs). Results siRNA-TIMP-1 alleviated carbon tetrachloride-induced liver fibrosis, reducing macrophage migration and MCP-1 expression. Co-culturing macrophages with hepatic stellate cells (HSCs) post-TIMP-1 downregulation inhibited macrophage migration. In siRNA-TIMP-1-treated HSCs, microRNA-145 (miRNA-145) expression increased, while the expression of Friend leukemia virus integration-1 (Fli-1) and MCP-1 was inhibited. Downregulation of Fli-1 led to decreased MCP-1 expression, whereas Fli-1 overexpression increased MCP-1 expression within HSCs. Transfection with miRNA-145 mimics reduced the expression of both Fli-1 and MCP-1, while miRNA-145 inhibitors elevated the expression of both Fli-1 and MCP-1 in HSCs. miRNA-145 bound directly to the 3'-UTR of Fli-1, and miRNA-145-enriched EVs secreted by HSCs after TIMP-1 downregulation influenced macrophage recruitment. Conclusions TIMP-1 induces Fli-1 expression through miRNA-145, subsequently increasing MCP-1 expression and macrophage recruitment. MiRNA-145-enriched EVs from HSCs can transmit biological information and magnify the function of TIMP-1.
Collapse
Affiliation(s)
- Xiaoli Huang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xiaofan Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Yanhong Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
- Dongying People's Hospital, Dongying, Shandong, China
| | - Shuangjun Shen
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xu Fan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| |
Collapse
|
7
|
Fortuna V, Lima J, Oliveira GF, Oliveira YS, Getachew B, Nekhai S, Aschner M, Tizabi Y. Ferroptosis as an emerging target in sickle cell disease. Curr Res Toxicol 2024; 7:100181. [PMID: 39021403 PMCID: PMC11252799 DOI: 10.1016/j.crtox.2024.100181] [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: 12/30/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Sickle cell disease (SCD) is an inherited hemoglobin disorder marked by red blood cell sickling, resulting in severe anemia, painful episodes, extensive organ damage, and shortened life expectancy. In SCD, increased iron levels can trigger ferroptosis, a specific type of cell death characterized by reactive oxygen species (ROS) and lipid peroxide accumulation, leading to damage and organ impairments. The intricate interplay between iron, ferroptosis, inflammation, and oxidative stress in SCD underscores the necessity of thoroughly understanding these processes for the development of innovative therapeutic strategies. This review highlights the importance of balancing the complex interactions among various factors and exploitation of the knowledge in developing novel therapeutics for this devastating disease.
Collapse
Affiliation(s)
- Vitor Fortuna
- Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Jaqueline Lima
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Gabriel F. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Yasmin S. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Sergei Nekhai
- Center for Sickle Cell Disease, Departments of Microbiology and Medicine, Howard University College of Medicine, Washington, DC, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
8
|
Song C, Li D, Huang L, Zhang J, Zhao X. Role of Ferroptosis Regulation by Nrf2/NQO1 Pathway in Alcohol-Induced Cardiotoxicity In Vitro and In Vivo. Chem Res Toxicol 2024; 37:1044-1052. [PMID: 38833663 DOI: 10.1021/acs.chemrestox.4c00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The aim of the present study was to evaluate the cardiotoxic effects of alcohol and its potential toxic mechanism on ferroptosis in mice and H9c2 cells. Mice were intragastrically treated with three different concentrations of alcohol, 7, 14, and 28%, each day for 14 days. Body weight and electrocardiography (ECG) were recorded over the 14 day period. Serum creatine kinase (CK), lactic dehydrogenase (LDH), MDA, tissue iron, and GSH levels were measured. Cardiac tissues were examined histologically, and ferroptosis was assessed. In H9c2 cardiomyocytes, cell viability, reactive oxygen species (ROS), labile iron pool (LIP), and mitochondrial membrane potential (MMP) were measured. The proteins of ferroptosis were evaluated by the western blot technique in vivo and in vitro. The results showed that serum CK, LDH, MDA, and tissue iron levels significantly increased in the alcohol treatment group in a dose-dependent manner. The content of GSH decreased after alcohol treatment. ECG and histological examinations showed that alcohol impaired cardiac function and structure. In addition, the levels of ROS and LIP increased, and MMP levels decreased after alcohol treatment. Ferrostatin-1 (Fer-1) protected cells from lipid peroxidation. Western blotting analysis showed that alcohol downregulated the expression of Nrf2, NQO1, HO-1, and GPX4. The expressions of P53 and TfR were upregulated in vivo and in vitro. Fer-1 significantly alleviated alcohol-induced ferroptosis. In conclusion, the study showed that Nrf2/NQO1-dependent ferroptosis played a vital role in the cardiotoxicity induced by alcohol.
Collapse
Affiliation(s)
- Chunpu Song
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Dongjie Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Ling Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Jie Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Xiaoyan Zhao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
9
|
Zhao Y, Zhao C, Deng Y, Pan M, Mo G, Liao Z, Zhang X, Zhang D, Li H. PMAIP1 promotes J subgroup avian leukosis virus replication by regulating mitochondrial function. Poult Sci 2024; 103:103617. [PMID: 38547674 PMCID: PMC11180372 DOI: 10.1016/j.psj.2024.103617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 06/05/2024] Open
Abstract
Avian leukosis virus Subgroup J (ALV-J) exhibits high morbidity and pathogenicity, affecting approximately 20% of poultry farms. It induces neoplastic diseases and immunosuppression. Phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1), a proapoptotic mitochondrial protein in the B-cell lymphoma-2 (Bcl-2) family, plays a role in apoptosis in cancer cells. However, the connection between the PMAIP1 gene and ALV-J pathogenicity remains unexplored. This study investigates the potential impact of the PMAIP1 gene on ALV-J replication and its regulatory mechanisms. Initially, we examined PMAIP1 expression using quantitative real-time PCR (qRT-PCR) in vitro and in vivo. Furthermore, we manipulated PMAIP1 expression in chicken fibroblast cells (DF-1) and assessed its effects on ALV-J infection through qRT-PCR, immunofluorescence assay (IFA), and western blotting (WB). Our findings reveal a significant down-regulation of PMAIP1 in the spleen, lung, and kidney, coupled with an up-regulation in the bursa and liver of ALV-J infected chickens compared to uninfected ones. Additionally, DF-1 cells infected with ALV-J displayed a notable up-regulation of PMAIP1 at 6, 12, 24, 48, 74, and 108 h. Over-expression of PMAIP1 enhanced ALV-J replication, interferon expression, and proinflammatory factors. Conversely, interference led to contrasting results. Furthermore, we observed that PMAIP1 promotes virus replication by modulating mitochondrial function. In conclusion, the PMAIP1 gene facilitates virus replication by regulating mitochondrial function, thereby enriching our understanding of mitochondria-related genes and their involvement in ALV-J infection, offering valuable insights for avian leukosis disease resistance strategies.
Collapse
Affiliation(s)
- Yongxia Zhao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, 510642 China
| | - Changbin Zhao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, 510642 China
| | - Yuelin Deng
- Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, 510642 China; Department of Animal Nutrition System, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Ming Pan
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, 510642 China
| | - Guodong Mo
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, 510642 China
| | - Zhiying Liao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, 510642 China
| | - Xiquan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, 510642 China
| | - Dexiang Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, 510642 China
| | - Hongmei Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, 510642 China.
| |
Collapse
|
10
|
Huang Y, Liu G, Zheng F, Chen J, Lin Y, Wang J, Huang Y, Peng Y. Asymmetric silicon phthalocyanine based nanoparticle with spatiotemporally targeting of mitochondria for synergistic apoptosis-ferroptosis antitumor treatment. Colloids Surf B Biointerfaces 2024; 238:113890. [PMID: 38608462 DOI: 10.1016/j.colsurfb.2024.113890] [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: 12/19/2023] [Revised: 03/20/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024]
Abstract
A promising therapeutic strategy in cancer treatment merges photodynamic therapy (PDT) induced apoptosis with ferroptosis, a form of programmed cell death governed by iron-dependent lipid peroxidation. Given the pivotal role of mitochondria in ferroptosis, the development of photosensitizers that specifically provoke mitochondrial dysfunction and consequentially trigger ferroptosis via PDT is of significant interest. To this end, we have designed and synthesized a novel nanoparticle, termed FECTPN, tailored to address this requisite. FECTPN harnesses a trifecta of critical attributes: precision mitochondria targeting, photoactivation capability, pH-responsive drug release, and synergistic apoptosis-ferroptosis antitumor treatment. This nanoparticle was formulated by conjugating an asymmetric silicon phthalocyanine, Chol-SiPc-TPP, with the ferroptosis inducer Erastin onto a ferritin. The Chol-SiPc-TPP is a chemically crafted entity featuring cholesteryl (Chol) and triphenylphosphine (TPP) functionalities bonded axially to the silicon phthalocyanine, enhancing mitochondrial affinity and leading to effective PDT and subsequent apoptosis of cells. Upon cellular uptake, FECTPN preferentially localizes to mitochondria, facilitated by Chol-SiPc-TPP's targeting mechanics. Photoactivation induces the synchronized release of Chol-SiPc-TPP and Erastin in the mitochondria's alkaline domain, driving the escalation of both ROSs and lipid peroxidation. These processes culminate in elevated antitumor activity compared to the singular application of Chol-SiPc-TPP-mediated PDT. A notable observation is the pronounced enhancement in glutathione peroxidase-4 (GPX4) expression within MCF-7 cells treated with FECTPN and subjected to light exposure, reflecting intensified oxidative stress. This study offers compelling evidence that FECTPN can effectively induce ferroptosis and reinforces the paradigm of a synergistic apoptosis-ferroptosis pathway in cancer therapy, proposing a novel route for augmented antitumor treatments.
Collapse
Affiliation(s)
- Yan Huang
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou, China
| | - Guowei Liu
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou, China
| | - Fangmei Zheng
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou, China
| | - Jianling Chen
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Provincial Key Laboratory for Photonics Technology, Institute of Laser and Optoelectronics Technology, Fujian Normal University, Fuzhou, China
| | - Youyu Lin
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Junwei Wang
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou, China
| | - Yide Huang
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou, China.
| | - Yiru Peng
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou, China.
| |
Collapse
|
11
|
Zhou X, Li Y, Zhang X, Li B, Jin S, Wu M, Zhou X, Dong Q, Du J, Zhai W, Wu Y, Qiu L, Li G, Qi Y, Zhao W, Gao Y. Hemin blocks TIGIT/PVR interaction and induces ferroptosis to elicit synergistic effects of cancer immunotherapy. SCIENCE CHINA. LIFE SCIENCES 2024; 67:996-1009. [PMID: 38324132 DOI: 10.1007/s11427-023-2472-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/23/2023] [Indexed: 02/08/2024]
Abstract
The immune checkpoint TIGIT/PVR blockade exhibits significant antitumor effects through activation of NK and CD8+ T cell-mediated cytotoxicity. Immune checkpoint blockade (ICB) could induce tumor ferroptosis through IFN-γ released by immune cells, indicating the synergetic effects of ICB with ferroptosis in inhibiting tumor growth. However, the development of TIGIT/PVR inhibitors with ferroptosis-inducing effects has not been explored yet. In this study, the small molecule Hemin that could bind with TIGIT to block TIGIT/PVR interaction was screened by virtual molecular docking and cell-based blocking assay. Hemin could effectively restore the IL-2 secretion from Jurkat-hTIGIT cells. Hemin reinvigorated the function of CD8+ T cells to secrete IFN-γ and the elevated IFN-γ could synergize with Hemin to induce ferroptosis in tumor cells. Hemin inhibited tumor growth by boosting CD8+ T cell immune response and inducing ferroptosis in CT26 tumor model. More importantly, Hemin in combination with PD-1/PD-L1 blockade exhibited more effective antitumor efficacy in anti-PD-1 resistant B16 tumor model. In summary, our finding indicated that Hemin blocked TIGIT/PVR interaction and induced tumor cell ferroptosis, which provided a new therapeutic strategy to combine immunotherapy and ferroptosis for cancer treatment.
Collapse
Affiliation(s)
- Xiaowen Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yang Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiangrui Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Beibei Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Shengzhe Jin
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Menghan Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiuman Zhou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen Campus, Shenzhen, 518107, China
| | - Qingyu Dong
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiangfeng Du
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lu Qiu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen Campus, Shenzhen, 518107, China
| | - Guodong Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen Campus, Shenzhen, 518107, China.
| |
Collapse
|
12
|
Zhang N, Yang P, Li Y, Ouyang Q, Hou F, Zhu G, Zhang B, Huang J, Jia J, Xu A. Serum Iron Overload Activates the SMAD Pathway and Hepcidin Expression of Hepatocytes via SMURF1. J Clin Transl Hepatol 2024; 12:227-235. [PMID: 38426189 PMCID: PMC10899870 DOI: 10.14218/jcth.2023.00440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
Background and Aims Liver iron overload can induce hepatic expression of bone morphogenic protein (BMP) 6 and activate the BMP/SMAD pathway. However, serum iron overload can also activate SMAD but does not induce BMP6 expression. Therefore, the mechanisms through which serum iron overload activates the BMP/SMAD pathway remain unclear. This study aimed to clarify the role of SMURF1 in serum iron overload and the BMP/SMAD pathway. Methods A cell model of serum iron overload was established by treating hepatocytes with 2 mg/mL of holo-transferrin (Holo-Tf). A serum iron overload mouse model and a liver iron overload mouse model were established by intraperitoneally injecting 10 mg of Holo-Tf into C57BL/6 mice and administering a high-iron diet for 1 week followed by a low-iron diet for 2 days. Western blotting and real-time PCR were performed to evaluate the activation of the BMP/SMAD pathway and the expression of hepcidin. Results Holo-Tf augmented the sensitivity and responsiveness of hepatocytes to BMP6. The E3 ubiquitin-protein ligase SMURF1 mediated Holo-Tf-induced SMAD1/5 activation and hepcidin expression; specifically, SMURF1 expression dramatically decreased when the serum iron concentration was increased. Additionally, the expression of SMURF1 substrates, which are important molecules involved in the transduction of BMP/SMAD signaling, was significantly upregulated. Furthermore, in vivo analyses confirmed that SMURF1 specifically regulated the BMP/SMAD pathway during serum iron overload. Conclusions SMURF1 can specifically regulate the BMP/SMAD pathway by augmenting the responsiveness of hepatocytes to BMPs during serum iron overload.
Collapse
Affiliation(s)
- Ning Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Gastroenterology, Beijing Shunyi Hospital, Beijing, China
| | - Pengyao Yang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanmeng Li
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qin Ouyang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Fei Hou
- Department of Critical Liver Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guixin Zhu
- MOE Key Laboratory of Protein Science, School of Life Sciences, Tsinghua University, Beijing, China
| | - Bei Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jian Huang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Anjian Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Zhang C, Zhai T, Zhu J, Wei D, Ren S, Yang Y, Gao F, Zhao L. Research Progress of Antioxidants in Oxidative Stress Therapy after Spinal Cord Injury. Neurochem Res 2023; 48:3473-3484. [PMID: 37526867 DOI: 10.1007/s11064-023-03993-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/08/2023] [Accepted: 07/09/2023] [Indexed: 08/02/2023]
Abstract
Spinal cord injury (SCI) is a serious problem in the central nervous system resulting in high disability and mortality with complex pathophysiological mechanisms. Oxidative stress is one of the main secondary reactions of SCI, and its main pathophysiological marker is the production of excess reactive oxygen species. The overproduction of reactive oxygen species and insufficient antioxidant capacity lead to the occurrence of oxidative stress and neuroinflammation, and the dysregulation of oxidative stress and neuroinflammation leads to further aggravation of damage. Oxidative stress can initiate a variety of inflammatory and apoptotic pathways, and targeted antioxidant therapy can greatly reduce oxidative stress and reduce neuroinflammation, which has a certain positive effect on rehabilitation and prognosis in SCI. This article reviewed the research on different types of antioxidants and related treatments in SCI, focusing on the mechanisms of oxidative stress.
Collapse
Affiliation(s)
- Can Zhang
- Medical School of Yan'an University, No. 580 Shengdi Road, Baota District, Yan'an, 716000, Shaanxi, China
| | - Tianyu Zhai
- Medical School of Yan'an University, No. 580 Shengdi Road, Baota District, Yan'an, 716000, Shaanxi, China
| | - Jinghui Zhu
- Medical School of Yan'an University, No. 580 Shengdi Road, Baota District, Yan'an, 716000, Shaanxi, China
| | - Dongmin Wei
- Medical School of Yan'an University, No. 580 Shengdi Road, Baota District, Yan'an, 716000, Shaanxi, China
| | - Shuting Ren
- Medical School of Yan'an University, No. 580 Shengdi Road, Baota District, Yan'an, 716000, Shaanxi, China
| | - Yanling Yang
- Medical School of Yan'an University, No. 580 Shengdi Road, Baota District, Yan'an, 716000, Shaanxi, China
| | - Feng Gao
- Medical School of Yan'an University, No. 580 Shengdi Road, Baota District, Yan'an, 716000, Shaanxi, China
| | - Lin Zhao
- Medical School of Yan'an University, No. 580 Shengdi Road, Baota District, Yan'an, 716000, Shaanxi, China.
| |
Collapse
|
14
|
Xiao L, Tang R, Wang J, Wan D, Yin Y, Xie L. Gut microbiota bridges the iron homeostasis and host health. SCIENCE CHINA. LIFE SCIENCES 2023; 66:1952-1975. [PMID: 37515687 DOI: 10.1007/s11427-022-2302-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/08/2023] [Indexed: 07/31/2023]
Abstract
The gut microbiota acts as a symbiotic microecosystem that plays an indispensable role in the regulation of a number of metabolic processes in the host by secreting secondary metabolites and impacting the physiology and pathophysiology of numerous organs and tissues through the circulatory system. This relationship, referred to as the "gut-X axis", is associated with the development and progression of disorders, including obesity, fatty liver and Parkinson's disease. Given its importance, the gut flora is a vital research area for the understanding and development of the novel therapeutic approaches for multiple disorders. Iron is a common but necessary element required by both mammals and bacteria. As a result, iron metabolism is closely intertwined with the gut microbiota. The host's iron homeostasis affects the composition of the gut microbiota and the interaction between host and gut microbiota through various mechanisms such as nutrient homeostasis, intestinal peaceability, gut immunity, and oxidative stress. Therefore, understanding the relationship between gut microbes and host iron metabolism is not only of enormous significance to host health but also may offer preventative and therapeutic approaches for a number of disorders that impact both parties. In this review, we delve into the connection between the dysregulation of iron metabolism and dysbiosis of gut microbiota, and how it contributes to the onset and progression of metabolic and chronic diseases.
Collapse
Affiliation(s)
- Lanling Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Rui Tang
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China
| | - Jie Wang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
- Department of Stomatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, China.
| |
Collapse
|
15
|
Zhang Q, Ding H, Yu X, Wang Q, Li X, Zhang R, Feng J. Plasma non-transferrin-bound iron uptake by the small intestine leads to intestinal injury and intestinal flora dysbiosis in an iron overload mouse model and Caco-2 cells. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2041-2055. [PMID: 37452897 DOI: 10.1007/s11427-022-2347-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/27/2023] [Indexed: 07/18/2023]
Abstract
Iron overload often occurs during blood transfusion and iron supplementation, resulting in the presence of non-transferrin-bound iron (NTBI) in host plasma and damage to multiple organs, but effects on the intestine have rarely been reported. In this study, an iron overload mouse model with plasma NTBI was established by intraperitoneal injection of iron dextran. We found that plasma NTBI damaged intestinal morphology, caused intestinal oxidative stress injury and reactive oxygen species (ROS) accumulation, and induced intestinal epithelial cell apoptosis. In addition, plasma NTBI increased the relative abundance of Ileibacterium and Desulfovibrio in the cecum, while the relative abundance of Faecalibaculum and Romboutsia was reduced. Ileibacterium may be a potential microbial biomarker of plasma NTBI. Based on the function prediction analysis, plasma NTBI led to the weakening of intestinal microbiota function, significantly reducing the function of the extracellular structure. Further investigation into the mechanism of injury showed that iron absorption in the small intestine significantly increased in the iron group. Caco-2 cell monolayers were used as a model of the intestinal epithelium to study the mechanism of iron transport. By adding ferric ammonium citrate (FAC, plasma NTBI in physiological form) to the basolateral side, the apparent permeability coefficient (Papp) values from the basolateral to the apical side were greater than 3×10-6 cm s-1. Intracellular ferritin level and apical iron concentration significantly increased, and SLC39A8 (ZIP8) and SLC39A14 (ZIP14) were highly expressed in the FAC group. Short hairpin RNA (shRNA) was used to knock down ZIP8 and ZIP14 in Caco-2 cells. Transfection with ZIP14-specific shRNA decreased intracellular ferritin level and inhibited iron uptake. These results revealed that plasma NTBI may cause intestinal injury and intestinal flora dysbiosis due to the uptake of plasma NTBI from the basolateral side into the small intestine, which is probably mediated by ZIP14.
Collapse
Affiliation(s)
- Qian Zhang
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Haoxuan Ding
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaonan Yu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qiwen Wang
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xuejiao Li
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ruiqiang Zhang
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jie Feng
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|