1
|
Liu Y, Huang J, Luo J. IGHG4: innovative diagnostic biomarkers for iron overload in β-thalassemia patients. Hematology 2025; 30:2433154. [PMID: 39676317 DOI: 10.1080/16078454.2024.2433154] [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: 07/28/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
OBJECTIVES This study aims to investigate the serotransferrin (TF), complement C1s subcomponent (C1S), immunoglobulin heavy constant gamma 4 (IGHG4), hemoglobin subunit alpha (HBA1), and clusterin (CLU) contents in β-thalassemia patients, and explores their physiological role as potential non-invasive bioindicators for disease diagnosis and iron overload. METHODS A total of 62 children with β-thalassemia were recruited and categorized by genotype, along with 17 healthy pediatric volunteers for analysis. The circulating ferritin content was evaluated, and plasma levels of TF, C1S, IGHG4, HBA1, and CLU were assessed using ELISA. The primary outcome of this study was the correlation between the five protein marker levels and iron overload. Continuous variables were analyzed using the Student's t-test or the Mann-Whitney U test. A binary logistic regression model identified independent predictors of iron overload in patients with β-thalassemia. Receiver operating characteristics (ROC) were employed to evaluate the model's performance. RESULTS The IGHG4 protein content was significantly lower in β-thalassemia patients compared to healthy controls. The IGHG4 protein content was reduced in the β+/β0 and β0/β0 patient populations compared to controls, with no significant difference observed between the β+/β0 group and healthy controls. A strong inverse relationship was identified between the IGHG4 protein content and SF concentration (r = -0.322, p = 0.004). Finally, plasma IGHG4 levels demonstrated adequate diagnostic capability, as indicated by our ROC curve analysis. CONCLUSION In conclusion, decreased IGHG4 protein levels are significantly associated with the degree of iron overload in β-thalassemia patients and may serve as a possible biomarker for evaluating iron overload.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Jinfang Huang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Jianming Luo
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| |
Collapse
|
2
|
Shaohui W, Xiangzhen K, Jingyi Y, Jindong T, Ye T, Xueguang L, Bo C, Yijie L, Jiucun W, Xinyu L, Jingdong T, Shuai J. STEAP3 alleviates inflammation and fibrosis via iron metabolism in ischemia/reperfusion-associated lung injury. Biochim Biophys Acta Mol Basis Dis 2025:167912. [PMID: 40412729 DOI: 10.1016/j.bbadis.2025.167912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 05/14/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Lower limb ischemia/reperfusion (LIRI)-associated lung injury is a common complication observed in patients with lower extremity occlusion and prolonged tourniquet exposure, leading to significant morbidity and mortality. The lung is the primary remote organ affected by LIRI, owing to its high oxygen demands. Excessive iron accumulation has been implicated in pulmonary injury pathogenesis. Although STEAP3, a key regulator of iron metabolism, has been shown to play a role in various organ injuries and inflammation, but its involvement in LIRI-associated lung injury has not been explored. In this study, we investigated the role of STEAP3 in LIRI-induced lung injury using a mouse model of tourniquet-induced LIRI. Lung injury severity was assessed by wet/dry weight ratio, pro-inflammatory cytokines (IL-1β, IL-6), and the pro-fibrotic marker TGF-β1. Bulk RNA sequencing of lung tissues identified differentially expressed genes and pathways. In vitro, a hypoxia/re‑oxygenation (H/R) model and iron treatment were employed using HUVEC cells. The results showed significant lung edema, macrophage infiltration, and elevated inflammation in LIRI mice, accompanied by iron overload in endothelial cells. Pathway analysis revealed that differentially expressed genes were enriched in inflammatory response regulation and ferroptosis. Both in vivo and in vitro models exhibited activation of the NF-κB and TGFβ1/SMAD pathways, with STEAP3 expression reduced in LIRI and H/R conditions. Overexpression of STEAP3 reversed iron accumulation and inhibited NF-κB and TGFβ1/SMAD signaling. These findings suggest that excessive iron accumulation exacerbates LIRI-associated lung injury, and STEAP3 acts as a potential therapeutic target, offering protection by modulating inflammatory and fibrotic signaling pathways.
Collapse
Affiliation(s)
- Wang Shaohui
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Kong Xiangzhen
- Human Phenome Institute, and Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China
| | - Yang Jingyi
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Tong Jindong
- Department of Vascular Surgery, Shanghai Key Laboratory of Vascular Lesion Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Tian Ye
- Department of Vascular Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Lin Xueguang
- Department of Vascular Surgery, Shanghai Key Laboratory of Vascular Lesion Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Chen Bo
- Department of Vascular Surgery, Shanghai Key Laboratory of Vascular Lesion Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Lu Yijie
- Department of Vascular Surgery, Shanghai Key Laboratory of Vascular Lesion Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Wang Jiucun
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China; Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; Human Phenome Institute, and Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China
| | - Ling Xinyu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Tang Jingdong
- Department of Vascular Surgery, Shanghai Key Laboratory of Vascular Lesion Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| | - Jiang Shuai
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China; Human Phenome Institute, and Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China; Department of Vascular Surgery, Shanghai Key Laboratory of Vascular Lesion Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| |
Collapse
|
3
|
Zhang B, Chen Y, Gu X, Zheng Y, Jiang ZH. Fucosyltransferase 11 restrains ferroptosis via upregulation GPX4 expression in gastric cancer. BMC Cancer 2025; 25:923. [PMID: 40405129 PMCID: PMC12100908 DOI: 10.1186/s12885-025-14340-4] [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: 07/12/2024] [Accepted: 05/15/2025] [Indexed: 05/24/2025] Open
Abstract
Ferroptosis is a novel iron-dependent type of programmed cell death that is characterized by the oxidation of lipids by divalent iron ions to produce lipid peroxides, which leads to cell death. Fucosyltransferase 11 (FUT11) is highly expressed in most tumors and is involved in tumorigenesis. However, there have been few studies regarding the relationship between FUT11 and ferroptosis. In this study, we found that FUT11 expression was abnormally high in gastric cancer (GC) cells and that the prognosis of patients with GC and high expression of FUT11 was poor. FUT11 expression was significantly correlated with the TNM stage of GC.Specific knockdown of FUT11 significantly inhibited the proliferation of GC cells, reduced the abundance of the key anti-ferroptotic protein glutathione peroxidase 4(GPX4), induced lipid peroxidation and ferroptosis in GC cells, and inhibited the proliferation of these cells. The overexpression of GPX4 reduced the inhibitory effect of FUT11 on GC cells. In addition, the knockdown of FUT11 significantly inhibited GC tumor growth in mice, and this inhibitory effect was reduced by the overexpression of GPX4. In conclusion, we have shown that FUT11 promotes GC progression by targeting GPX4, thereby inhibiting ferroptosis in GC cells. These findings suggest that FUT11 is a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Bingbing Zhang
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, The Yancheng Clinical Medical College of Jiangsu University, Yancheng, Jiangsu, 224006, China
| | - Yali Chen
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, The Yancheng Clinical Medical College of Jiangsu University, Yancheng, Jiangsu, 224006, China
| | - Xuezhou Gu
- Department of General Surgery, Sheyang People's Hospital, Yancheng, Jiangsu, 224006, China
| | - Yu Zheng
- Department of Laboratory Medicine, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, The Yancheng Clinical Medical College of Jiangsu University, Yancheng, Jiangsu, 224006, China
| | - Zhong Hua Jiang
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, The Yancheng Clinical Medical College of Jiangsu University, Yancheng, Jiangsu, 224006, China.
| |
Collapse
|
4
|
Tata P, Ghosh A, Jamma T, Kulkarni O, Ganesan R, Ray Dutta J. Caffeic Acid-Biogenic Amine Complexes Outperform Standard Drugs in Reducing Toxicity: Insights from In Vivo Iron Chelation Studies. Mol Pharm 2025. [PMID: 40315048 DOI: 10.1021/acs.molpharmaceut.4c01424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Iron homeostasis imbalance, caused by conditions such as thalassemia, sickle cell anemia, and myocardial infarction, often results in elevated free iron levels, leading to ferroptosis and severe organ damage. While current iron chelators like deferoxamine (DFO) and deferiprone are effective, they are associated with significant side effects, including nephrotoxicity, gastrointestinal bleeding, and liver fibrosis. This creates an urgent need for safer, natural-product-based alternatives for effective iron chelation therapy (ICT). This study investigates caffeic acid (CA)-based complexes with biogenic amines, specifically spermine (CA-Sp) and histidine (CA-His), as potential ICT candidates. Initial in vitro assays on HEK-293 cells under iron dextran (ID)-induced toxicity have demonstrated their protective effects, with CA-Sp exhibiting superior efficacy. The in vivo studies in mice have further validated their potential, showing remarkable iron chelation and toxicity mitigation compared to DFO. Inductively coupled plasma mass spectrometry (ICP-MS) reveals significant iron excretion in fecal matter in the treatment group along with reductions in serum ferritin levels. The markers of nephrotoxicity (creatinine) and liver function (ALT, AST) have also been shown to be normalized in treated groups, while immunological analyses have revealed restored levels of neutrophils, T cells, and B cells. Additionally, the inflammatory cytokines, TNF-α and IL-6, have exhibited significant reductions, with the CA-based formulations surpassing the effects of DFO. Histological analyses using Prussian blue staining have further confirmed reduced iron deposition in vital organs such as the liver, kidney, and spleen. These findings highlight CA-Sp as a particularly promising candidate for ICT, offering a safer and more effective strategy for managing iron overload and its associated complications.
Collapse
Affiliation(s)
- Pranathi Tata
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Hyderabad, Telangana 500078,India
| | - Aparajita Ghosh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Hyderabad, Telangana 500078,India
| | - Trinath Jamma
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Hyderabad, Telangana 500078,India
| | - Onkar Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Hyderabad, Telangana 500078,India
| | - Ramakrishnan Ganesan
- Department of Chemistry, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Hyderabad, Telangana 500078,India
| | - Jayati Ray Dutta
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Hyderabad, Telangana 500078,India
| |
Collapse
|
5
|
Xiao W, Yike W, Gongwen L, Youjia X. Ferroptosis-mediated immune responses in osteoporosis. J Orthop Translat 2025; 52:116-125. [PMID: 40271049 PMCID: PMC12017889 DOI: 10.1016/j.jot.2025.03.011] [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: 12/08/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Osteoporosis is a common systemic metabolic disease, characterized by decreased bone mass and susceptibility to fragility fractures, often associated with aging, menopause, genetics, and immunity. Ferroptosis plays an underestimated yet crucial role in the further impact of immune function changes on osteoporosis. Cell ferroptosis can induce alterations in immune function, subsequently influencing bone metabolism. In this context, this review summarizes several mechanisms of ferroptosis and introduces the latest insights on how ferroptosis regulates immune responses, exploring the interactions between ferroptosis and other mechanisms such as oxidative stress, inflammation, etc. This review elucidates potential treatment strategies for osteoporosis, emphasizing the promising potential of ferroptosis as an emerging target in the treatment of osteoporosis. In conclusion, preparations related to ferroptosis exhibit substantial clinical promise for enhancing bone mass restoration. The translational potential of this article: This review elucidates a nuanced conversation between the immune system and osteoporosis, with ferroptosis serving as the connecting link. These findings underscore the potential of ferroptosis inhibition as a therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Wang Xiao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wang Yike
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liu Gongwen
- Department of Orthopaedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xu Youjia
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
6
|
Qin Y, Zhou R. Causal associations between iron deficiency anemia and digestive system cancers: evidence from a bidirectional two-sample Mendelian randomization study. Discov Oncol 2025; 16:650. [PMID: 40310589 PMCID: PMC12045922 DOI: 10.1007/s12672-025-02367-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND & AIMS Iron deficiency anemia (IDA) is associated with digestive system cancers (DSCs), but the causal relationship is poorly understood. This two-sample bidirectional Mendelian randomization (MR) study investigated the causal association between IDA and five types of DSCs. METHODS This study pooled data from a genome-wide association study of IDA (6,087 cases and 211,115 controls of European ancestry) and DSCs. IVW, weighted median, weighted mode, and MR-Egger regression were used to assess causal associations between IDA and DSCs. Sensitivity analysis included Cochran's Q test for heterogeneity, MR-PRESSO for pleiotropy, and leave-one-out method for robustness. RESULTS The MR analysis used 12 single-nucleotide polymorphisms (SNPs) associated with IDA as instrumental variables (IVs). In contrast, the reverse MR analysis used 20 SNPs associated with the five types of DSC as IVs. Genetic predictions revealed no significant association between IDA and the risk of DSCs: (odds ratio [OR]: 1.00; 95% confidence interval [CI] [0.76, 1.31]; P = 0.979), esophageal (OR: 0.94; 95% CI [0.67, 1.31]; P = 0.699), pancreatic (OR: 1.14; 95% CI [0.68, 1.92]; P = 0.615), liver (OR: 1.12; 95% CI [0.51, 2.47]; P = 0.776), and stomach (OR: 1.04; 95% CI [0.71, 1.54]; P = 0.830) cancers. Reverse MR also indicated no causal association between DSC and IDA. MR-Egger regression showed minimal heterogeneity impact except for colorectal cancer (heterogeneity P = 0.002). MR-PRESSO identified no outliers. CONCLUSION The present MR analysis shows no causal associations between IDA and the risk of DSCs.
Collapse
Affiliation(s)
- Yan Qin
- Department of Gastroenterology, Changzhou Maternal and Child Health Care Hospital, Changzhou, 213000, China.
| | - Rong Zhou
- Respiratory and Critical Care Medicine Department, The First People's Hospital of Changzhou, Changzhou, 213000, China
| |
Collapse
|
7
|
Wang D, Zhou J, Sun X, Niu X. The Essence of Nature Can be the Simplest (3) Holistic Energy: Extracellular Fenton Reactions of All Cells. Chem Biodivers 2025:e202500942. [PMID: 40263106 DOI: 10.1002/cbdv.202500942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 04/24/2025]
Abstract
Cooperative mechanisms are widely regarded as pivotal drivers in the evolutionary transition from unicellular to multicellular life. However, the mechanism of how multicellular organisms integrate the individual energy of all cells into holistic energy and how exactly the holistic energy is manifested remains poorly understood. Traditionally, relying on ATP within heart muscle cells to drive a permanent heartbeat (called cardiac contraction) is responsible for the vital holistic biological process of transporting and distributing oxygen and nutrients to cells throughout the body, but the limited ATP reserve in cardiac muscle challenges this paradigm. Recent studies suggest that cellular energy production involves dual pathways: canonical ATP synthesis and extracellular Fenton reactions. Here, we propose a novel paradigm wherein the circulatory system serves as a collective platform for integrating extracellular Fenton reaction capabilities of all cells, generating sustained and powerful energy to drive the heartbeat and maintain body temperature. This cooperative mechanism for holistic energy aligns with Bergmann's rule and the holistic principles of traditional Chinese medicine (TCM). Holistic energy in circulatory system supports pulse diagnosis that evaluates systemic health through vascular dynamics. Furthermore, the interaction and balance between holistic energy and individual energy can also explain the rationality of tumor occurrence.
Collapse
Affiliation(s)
- Donglou Wang
- School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources & Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, P. R. China
| | - Jiao Zhou
- School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources & Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, P. R. China
| | - Xingrong Sun
- School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources & Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, P. R. China
| | - Xuemei Niu
- School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources & Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, P. R. China
| |
Collapse
|
8
|
Zhang L, Xu C, Li M, Lu X, Sheng Y, Chen L, Shen J, Miao Y, Xiao Z, Liu N, Zhang Y, Chen Q, Wang Y. Gambogic Acid Based Coordination Polymer Reinforces High-Intensity Focused Ultrasound Treatment of Gynecologic Malignancies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501664. [PMID: 40223396 DOI: 10.1002/adma.202501664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/26/2025] [Indexed: 04/15/2025]
Abstract
High-intensity focused ultrasound (HIFU) is emerging as a promising non-invasive treatment for solid tumors. Nevertheless, HIFU may also induce the upregulation of Heat Shock Protein 90 (HSP-90), potentially resulting in resistance to HIFU. Besides, although it is effective against in situ tumors, challenges remain with tumor metastasis and recurrence. Herein, the innovative design of gambogic acid (GA) based coordination polymer-GAZn-PEG nanoparticles (GAZn-PEG NPs) are synthesized through the coordination of GA with zinc ions (Zn2+), and subsequently functionalized with lipid bilayer incorporating polyethylene glycol (PEG), sensitizing HIFU for the treatment of cervical and ovarian cancers. Briefly, under HIFU exposure, GA markedly suppresses the expression of HSP-90, thereby increasing the tumor's sensitivity to HIFU therapy. Furthermore, Zn2+ not only overcome the issue of GA's poor water solubility but also synergistically stimulate immune responses in conjunction with GA. More intriguingly, it has been discovered that GAZn-PEG can effectively activate the cyclic GMP-AMP synthase-stimulator of the interferon genes (cGAS-STING) pathway, thereby enhancing the immune responses provoked by HIFU. Specifically, GAZn-PEG NPs show a remarkable increase in dendritic cell activation and the effective stimulation of the cGAS-STING pathway, crucial for long-term protection against tumor recurrence and metastasis.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, 200030, China
- National Center for Translational Medicine (Shanghai) - Maternal and Children's Branch, Shanghai, 200030, China
| | - Chen Xu
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, 200030, China
- National Center for Translational Medicine (Shanghai) - Maternal and Children's Branch, Shanghai, 200030, China
| | - Mingzhuang Li
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices Soochow University, Suzhou, 215123, China
- Department of Gynecology, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Xiaojing Lu
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, 200030, China
- National Center for Translational Medicine (Shanghai) - Maternal and Children's Branch, Shanghai, 200030, China
| | - Yaru Sheng
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, 200030, China
- National Center for Translational Medicine (Shanghai) - Maternal and Children's Branch, Shanghai, 200030, China
| | - Linfu Chen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices Soochow University, Suzhou, 215123, China
| | - Jingjing Shen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices Soochow University, Suzhou, 215123, China
| | - Yu Miao
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices Soochow University, Suzhou, 215123, China
| | - Zhisheng Xiao
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices Soochow University, Suzhou, 215123, China
| | - Nanhui Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices Soochow University, Suzhou, 215123, China
| | - Yueming Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices Soochow University, Suzhou, 215123, China
- Department of Gynecology, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Qian Chen
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, 200030, China
| | - Yudong Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, 200030, China
- National Center for Translational Medicine (Shanghai) - Maternal and Children's Branch, Shanghai, 200030, China
| |
Collapse
|
9
|
Li T, Adams J, Zhu P, Zhang T, Tu F, Gravitte A, Zhang X, Liu L, Casteel J, Yakubenko V, Williams DL, Li C, Wang X. The role of heme in sepsis induced Kupffer cell PANoptosis and senescence. Cell Death Dis 2025; 16:284. [PMID: 40221420 PMCID: PMC11993645 DOI: 10.1038/s41419-025-07637-6] [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: 12/12/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
Elevated heme levels, a consequence of hemolysis, are strongly associated with increased susceptibility to bacterial infections and adverse sepsis outcomes, particularly in older populations. However, the underlying mechanisms remain poorly understood. Using a cecal ligation and puncture (CLP) model of sepsis, we demonstrate that elevated heme levels correlate with Kupffer cell loss, increased bacterial burden, and heightened mortality. Mechanistically, we identify mitochondrial damage as a key driver of heme- and bacterial-induced Kupffer cell PANoptosis, a form of cell death integrating pyroptosis, apoptosis, and necroptosis, as well as cellular senescence. Specifically, heme activates phospholipase C gamma (PLC-γ), facilitating the translocation of cleaved gasdermin D (c-GSDMD) to mitochondria, resulting in GSDMD pore formation, mitochondrial dysfunction, and the release of mitochondrial DNA (mtDNA) during bacterial infection. This mitochondrial damage amplifies PANoptosis and triggers the cGAS-STING signaling pathway, further driving immune senescence. Notably, PLC-γ inhibition significantly reduces mitochondrial damage, cell death, and senescence caused by heme and bacterial infection. Furthermore, we show that hemopexin, a heme scavenger, effectively mitigates sepsis-induced Kupffer cell death and senescence, enhances bacterial clearance, and improves survival outcomes in both young and aged mice. These findings establish mitochondrial damage as a central mediator of heme induced Kupffer cell loss and highlight PLC-γ inhibition and hemopexin administration as promising therapeutic strategies for combating sepsis associated immune dysfunction.
Collapse
Affiliation(s)
- Tingting Li
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Joseph Adams
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Peilin Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Tao Zhang
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Fei Tu
- UMPC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Amy Gravitte
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jared Casteel
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Valentin Yakubenko
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Center of Excellence in Inflammation, Infectious Disease, and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - David L Williams
- Center of Excellence in Inflammation, Infectious Disease, and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Chuanfu Li
- Center of Excellence in Inflammation, Infectious Disease, and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Xiaohui Wang
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
- Center of Excellence in Inflammation, Infectious Disease, and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
| |
Collapse
|
10
|
Xu TY, Zhao JX, Chen MY, Miao ZW, Li ZY, Chang YQ, Wang YS, Miao CY. Exploring METRNL as a novel biomarker in sepsis: diagnostic potential and secretion mechanism. J Intensive Care 2025; 13:19. [PMID: 40205457 PMCID: PMC11983927 DOI: 10.1186/s40560-025-00780-4] [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: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Sepsis is a life-threatening condition with a high mortality rate in intensive care unit (ICU). However, rapid and accurate diagnostic criteria are still lacking. This pilot study explored the role of METRNL as a novel biomarker for sepsis by focusing on its diagnostic potential and rapid secretion mechanism. METHODS METRNL levels were measured in cell and animal models of sepsis. Serum samples from 107 sepsis patients and 95 non-septic controls in ICU were collected. Diagnostic performance of METRNL, Procalcitonin (PCT) and C-reactive protein (CRP) were assessed using ROC analysis. Endothelial cell-specific Metrnl gene knockout mice (EC-Metrnl-/- mice) were used to identify the source of METRNL secretion. Chemical inhibitors and RNA interference were used to explore the secretion pathways. RESULTS In lipopolysaccharide (LPS)-induced cell and mouse models of sepsis, METRNL levels significantly increased in a dose- and time-dependent manner. Similarly, in the cecal ligation and puncture mouse models, serum METRNL levels were elevated over time and correlated with sepsis severity. In animals, serum METRNL increased within 1 h post-modeling, preceding PCT and CRP. Clinically, sepsis patients had significantly higher serum METRNL levels. ROC analysis showed area under the curves [95% confidence intervals] of 0.943 [0.91-0.975] for METRNL, 0.955 [0.929-0.981] for PCT and 0.873 [0.825-0.921] for CRP. At the optimal cutoff value, METRNL (91.6%) exhibited relatively greater diagnostic specificity than PCT (88.4%) and CRP (69.5%). EC-Metrnl-/- reduced majority of serum Metrnl levels in sepsis mouse models. Inhibition of the endoplasmic reticulum-Golgi (ER-Golgi) pathway through chemical inhibitors or RNA interference significantly reduced METRNL levels in the supernatant of sepsis cell models compared to control groups. Similar results were obtained with Toll-like receptor 4 (TLR4) and ERK inhibitors. CONCLUSIONS This pilot study demonstrates that METRNL is a novel potential biomarker for sepsis with diagnostic capability comparable to that of PCT. Serum METRNL rapidly increased during the early phase of sepsis. Mechanistically, it mainly originates from the endothelium during sepsis, and TLR4-ERK signaling mediates the rapid secretion of METRNL via the classical ER-Golgi pathway in response to LPS stimulation.
Collapse
Affiliation(s)
- Tian-Ying Xu
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, China.
- Department of Anesthetic Pharmacology, School of Anesthesiology, Second Military Medical University/Naval Medical University, Shanghai, China.
| | - Jing-Xin Zhao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, China
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Ming-Yao Chen
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, China
- Department of Anesthetic Pharmacology, School of Anesthesiology, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Zhu-Wei Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Zhi-Yong Li
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Yong-Qing Chang
- Department of Critical Care Medicine, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Yu-Sheng Wang
- Department of Critical Care Medicine, Naval Medical Center of PLA, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, China.
| |
Collapse
|
11
|
Zheng N, Li D, Hu X, Yan L, Ding LY, Feng J, Ji T, He S, Huang Y, Hu J. Enhanced Sonodynamic Cancer Therapy through Boosting Reactive Oxygen Species and Depleting Glutathione. NANO LETTERS 2025; 25:5908-5915. [PMID: 40130822 DOI: 10.1021/acs.nanolett.5c00946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
The complex tumor microenvironment (TME) affects reactive oxygen species (ROS)-based therapies; breaking the limitations of the TME to enhance the effectiveness of sonodynamic therapy (SDT) is full of great challenges. Herein, iron atomically dispersed nanoparticles (Fe-N-C) were first reported as sonosensitizers with highly efficient ROS generation by overcoming TME limitations. Its peroxidase and catalase-like activities catalyze H2O2 to produce highly toxic ·OH and in situ O2, respectively, and then O2 molecules adsorbed at Fe active sites obviously lower the energy barrier for ·OH formation. Meanwhile, its glutathione-oxidase-like activity can rapidly consume glutathione (GSH) in the TME to induce tumor cell apoptosis and ferroptosis. Density functional theory calculation results elucidate the possible mechanism of ROS generation: O2 molecules are activated by receiving sonoelectrons to generate ·O2-, which further reacts with H2O to produce OH-. Then OH- is oxidized by sonoholes to form ·OH. Fe-N-C displays a superior tumor specificity SDT.
Collapse
Affiliation(s)
- Nannan Zheng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, P. R. China
| | - Dan Li
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, P. R. China
| | - Xin Hu
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, P. R. China
| | - Li Yan
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, P. R. China
| | - Ling-Yun Ding
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, P. R. China
| | - Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, P. R. China
| | - Tao Ji
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, P. R. China
| | - Shuqing He
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, P. R. China
| | - Yudai Huang
- State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources, College of Chemistry, Xinjiang University, Urumqi 830017, Xinjiang, P. R. China
| | - Junqing Hu
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, P. R. China
| |
Collapse
|
12
|
Lv J, Quan H, Lv J, Sui Y, Yu P, Guo S, Miao Y, Lv M. Argatroban and Menadione exert protective effects in ultraviolet-irradiated skin inflammation: A transcriptomic analysis based on identification of iron overload related biomarkers. Int Immunopharmacol 2025; 151:114334. [PMID: 40020462 DOI: 10.1016/j.intimp.2025.114334] [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: 11/07/2024] [Revised: 02/13/2025] [Accepted: 02/15/2025] [Indexed: 03/03/2025]
Abstract
Ultraviolet light (UV) can cause serious damage to human skin. The inflammatory reaction arising from repeated UV exposure leads to severe skin lesions and even promotes photo-carcinogenesis. Iron overload is featured by excessive iron intake and deposition and will promote inflammatory response inside cells. However, the core molecules involved in UV radiation induced iron overload and related anti-inflammatory strategies remain unclear. Signature genes involved in UV-irradiated skin were filtered through integrated datasets from the Gene Expression Omnibus (GEO) database. Subsequently, immune cell infiltration analysis was carried out to examine the relationship between signature gene expression and immune cell abundance. Single cell RNA-seq matrix data implicated in UV-irradiated skin was then applied to assess the expression level of signature genes in different cell clusters and to find out the core cell type and the key signaling pathway involved in UV radiation. Finally, cytological and animal experiments were conducted to investigate the potential of signature genes as therapeutic targets. SAT1 and RBMS1 were screened and validated as signature genes of UV irradiation. Immune cell infiltration analysis demonstrated that SAT1 and RBMS1 expression were associated closely with immune cell abundance, and skin fibroblasts were identified as the central cell type to communicate with other cell clusters in UV-irradiated skin. Disturbance of SAT1 exerted observably more suppressive effects on the release of inflammatory cytokines than overexpression of RBMS1. Two small molecule drugs targeting SAT1, namely Argatroban and Menadione, were predicted. Moreover, their therapeutic potentials in the treatment of UV-irradiated skin injury were confirmed experimentally.
Collapse
Affiliation(s)
- Jiacheng Lv
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Huilin Quan
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jiarui Lv
- Department of Organ Transplantation and Hepatobiliary, The First Hospital of China Medical University, Shenyang, China
| | - Yanan Sui
- Department of ophthalmology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Panpan Yu
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China.
| | - Yuwei Miao
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China.
| | - Mengzhu Lv
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
13
|
Wu J, Zheng Z, Wang J, Xiao W, Shi L, Fan L. Iron Status and Risk of Periodontitis and Dental Caries: A Mendelian Randomization Study. Int Dent J 2025; 75:1441-1449. [PMID: 39741062 PMCID: PMC11976561 DOI: 10.1016/j.identj.2024.11.018] [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: 07/30/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Previous studies have indicated a potential relationship between iron status and oral health outcomes, specifically periodontitis and dental caries. This study employed Mendelian randomization (MR) to investigate the causal effects of iron status on these oral health conditions. The focus of this study was on key iron biomarkers, namely serum iron, ferritin, transferrin saturation (TSAT), and total iron-binding capacity (TIBC). METHODS This two-sample MR analysis employed genome-wide association study (GWAS) data. The instrumental variables (IVs) were selected based on their genome-wide significance and independence from confounders. The statistical analyses employed the inverse variance weighted (IVW) method, MR-Egger regression, weighted median, and weighted mode. Additionally, sensitivity analyses were conducted to verify the reliability of the causal association results. RESULTS The MR analysis indicated a suggestive negative causal relationship between TIBC and periodontitis, with an odds ratios of 0.875 and a 95% CI of 0.766-0.998, with a P-value of .047. No significant other associations were found. The results of sensitivity analyses demonstrated the robustness of these findings. CONCLUSION This MR study suggested a potential negative association between TIBC and periodontitis, highlighting the importance of considering iron status in the clinical management of chronic periodontitis. However, more standardized, multi-population studies are needed to confirm this causality.
Collapse
Affiliation(s)
- Jiaqi Wu
- Department of Prosthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Institute of Stomatology, Southwest Medical University, Luzhou, China
| | - Ziyang Zheng
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Institute of Stomatology, Southwest Medical University, Luzhou, China
| | - Jinghan Wang
- Department of Prosthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Institute of Stomatology, Southwest Medical University, Luzhou, China
| | - Weiwei Xiao
- Department of Prosthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Institute of Stomatology, Southwest Medical University, Luzhou, China
| | - Liang Shi
- Department of Prosthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Institute of Stomatology, Southwest Medical University, Luzhou, China
| | - Liyuan Fan
- Department of Prosthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Institute of Stomatology, Southwest Medical University, Luzhou, China.
| |
Collapse
|
14
|
Yao Y, Chen Y, Fu J, Ding J, Zhou W, Chen X, Wan X. A metal-polyphenol network-based iron supplement with improved stability and reduced gastrointestinal toxicity for iron deficiency anemia therapy. Mater Today Bio 2025; 31:101598. [PMID: 40070867 PMCID: PMC11894331 DOI: 10.1016/j.mtbio.2025.101598] [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: 01/02/2025] [Revised: 02/05/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Iron deficiency anemia (IDA) is a global health concern, particularly affecting women and children of reproductive age. Although oral iron supplements are the standard treatment for IDA, their bioavailability is often compromised by food interactions, and they are associated with significant gastrointestinal side effects. To overcome these limitations, we developed a novel iron nano-supplement, TA-Fe NPs, based on metal-polyphenol networks (MPNs) formed through the coordination of tannic acid (TA) and Fe3+. These uniform nanoparticles (∼190 nm) offer enhanced chemical stability and reduced food interference compared to traditional iron supplements. The polyphenolic TA component provides antioxidant properties, effectively mitigating oxidative stress and inflammation induced by free iron ions. To further improve stability and intestinal absorption, TA-Fe NPs were encapsulated in an enteric coating (TA-Fe@L100) to protect against acidic conditions in the stomach. In a mouse model of IDA, TA-Fe@L100 demonstrated superior therapeutic efficacy compared to FeSO4, including improvements in hematological parameters, organ iron storage, and gut microbiota balance. Importantly, TA-Fe@L100 alleviated common gastrointestinal side effects associated with iron supplementation, presenting a promising alternative for IDA treatment. Our findings suggest that TA-Fe@L100 is a cost-effective and biocompatible oral iron supplement with minimal side effects, offering significant potential for broader clinical application in the management of IDA.
Collapse
Affiliation(s)
- Ying Yao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Yuanzheng Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Jie Fu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Xinyi Chen
- Yongkang First People's Hospital of Wenzhou Medical University, Jinhua, 321300, China
| | - Xiuping Wan
- Department of Gastroenterology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| |
Collapse
|
15
|
Ali I, Muhammad S, Naqvi SSZH, Wei L, Yan W, Khan MF, Mahmood A, Liu H, Shah W. Hepatitis B Virus-Associated Liver Carcinoma: The Role of Iron Metabolism and Its Modulation. J Viral Hepat 2025; 32:e14016. [PMID: 39445513 DOI: 10.1111/jvh.14016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/17/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Hepatitis B virus (HBV) infection is a significant contributor to the development of hepatocellular carcinoma (HCC), a leading cause of cancer-related mortality worldwide. Iron, a central co-factor in various metabolic pathways, plays an essential role in liver function, but its dysregulation can lead to severe health consequences. Accumulation of iron within hepatic cells over time is linked to increased liver injury and is strongly associated with sensitive exposure to a range of conditions, including cirrhosis, fibrosis and ultimately, HCC. This review explores the intricate interplay between iron metabolism and HCC within the context of HBV infection. Hepatic iron overload can arise from liver injury and disruptions in iron homeostasis, causing hepatic necrosis, inflammation, and fibrosis, ultimately culminating in carcinogenesis. Moreover, alterations in serum iron components in HBV-related scenarios have been observed to impact the persistence of HBV infection. Notably, the progression of HBV-associated liver damage exhibits distinct characteristics at various stages of liver disease. In addition to elucidating the complex relationship between iron metabolism and HCC in the context of HBV infection, this review also investigates the prognostic implications of systemic iron levels for HCC. Furthermore, it aims to provide a comprehensive understanding of the intricate interplay between iron metabolism and HCC, extending the discussion to the context of hepatitis C virus (HCV) infection. By shedding light on these multifaceted connections, this review aims to contribute to our understanding of the pathogenesis of HBV-associated HCC and potentially identify novel therapeutic avenues for intervention.
Collapse
Affiliation(s)
- Imran Ali
- Department of General Surgery, Subspecialty Hepatobiliary Surgery, Shanxi First Medical Hospital Affiliated With Shanxi Medical University, Yangzi Qu, Taiyuan, China
| | - Shoaib Muhammad
- Department of Urology, First Hospital of Shanxi Medical University, Yangzi Qu, Taiyuan, China
| | - Syed Shah Zaman Haider Naqvi
- Department of Endocrinology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences; Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lingxi Wei
- Laboratory of Physiology, Shanxi Medical University, Jing Zhong, China
| | - Wenqi Yan
- Shandong University, Ji Nan, Shandong, China
| | - Muhammad Fiaz Khan
- Department of Zoology, Hazara University, Mansehra, Khyber Pakhtunkhwa, Pakistan
| | - Ahmad Mahmood
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hong Liu
- Department of General Surgery, Subspecialty Hepatobiliary Surgery, Shanxi First Medical Hospital Affiliated With Shanxi Medical University, Yangzi Qu, Taiyuan, China
| | - Wahid Shah
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, China
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
16
|
Zhu L, Hu J, Wu X, Zhang J, Xu X, Huang X, Tian B, Zhao CX, Du Y, Wu L. Programmed enhancement of endogenous iron-mediated lysosomal membrane permeabilization for tumor ferroptosis/pyroptosis dual-induction. Nat Commun 2025; 16:3017. [PMID: 40148335 PMCID: PMC11950380 DOI: 10.1038/s41467-025-58124-7] [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: 09/11/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Ferroptosis and pyroptosis, as emerging regulated forms of cell death capable of overcoming apoptotic resistance, demonstrate promising potential in tumor therapy. Given that iron manipulation and reactive oxygen species elevation serve as common stimuli for both processes, inducing lysosomal membrane permeabilization (LMP) with ensuing release of lysosomal contents (including iron ions and cathepsins) is anticipated to realize dual induction of ferroptosis/pyroptosis. Herein, we report a folic acid and croconaine molecule-functionalized upconversion nanoparticle (UCNP-Cro/FA) that is able to mobilize intracellular stores of endogenous iron and spatiotemporally control the lysosome-intrinsic Fenton chemistry, thereby triggering LMP-associated cell death. The process of endogenous iron mobilization occurs through two key steps: Cro-mediated coordination of abundant Fe3+ ions within lysosomes, followed by UV-emitting upconversion core-mediated photoreduction, resulting in Fe2+ ions release. Both in vitro and in vivo experiments show that UCNP-Cro/FA + NIR treatment effectively boost LMP by endogenous iron-mediated •OH production, ultimately triggering irreversible tumor cell death via ferroptosis and Caspase-1/GSDMD-dependent pyroptosis pathways. Moreover, this process potentiates tumor immunogenicity, holding promise for tumor immunotherapy. Overall, this work proposes a feasible tumor therapy strategy that integrates ferroptosis and pyroptosis through the efficient application and activation of endogenous iron.
Collapse
Affiliation(s)
- Luwen Zhu
- Department of General Surgery, Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, P. R. China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jiahao Hu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xiaochuan Wu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jucong Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xinyi Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xiajie Huang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Bing Tian
- Institute of Biophysics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China.
| | - Chun-Xia Zhao
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA, Australia.
| | - Yongzhong Du
- Department of General Surgery, Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, P. R. China.
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China.
| | - Liming Wu
- Department of General Surgery, Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, P. R. China.
| |
Collapse
|
17
|
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
|
18
|
Zhou H, Fan Z, Da Y, Liu X, Wang C, Zhang T, Zhang J, Wu T, Liang J. Causal Relationships Between Iron Deficiency Anemia, Gut Microbiota, and Metabolites: Insights from Mendelian Randomization and In Vivo Data. Biomedicines 2025; 13:677. [PMID: 40149653 PMCID: PMC11940133 DOI: 10.3390/biomedicines13030677] [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: 02/05/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Iron deficiency anemia (IDA) is a common type of anemia in children and pregnant women. The effects of iron deficiency on gut microbiota and metabolic profiles are not fully understood. Methods: Mendelian randomization (MR) analysis was conducted to explore associations among IDA, gut microbiota, and metabolites. MR analysis was conducted using computational methods, utilizing human genetic data. Data were obtained from genome-wide association studies (GWAS), with inverse-variance-weighted (IVW) as the primary method. Animal models evaluated the effects of IDA on gut microbiota and metabolic profiles. Results: IVW analysis revealed significant associations between gut microbial taxa and IDA. The genus Desulfovibrio was protective (OR = 0.85, 95% CI: 0.77-0.93, p = 0.001), while Actinomyces (OR = 1.12, 95% CI: 1.01-1.23, p = 0.025) and family XIII (OR = 1.16, 95% CI: 1.01-1.32, p = 0.035) increased IDA risk. Glycine was protective (OR = 0.95, 95% CI: 0.91-0.99, p = 0.011), whereas medium low density lipoprotein (LDL) phospholipids increased risk (OR = 1.07, 95% CI: 1.00-1.15, p = 0.040). Animal models confirmed reduced Desulfovibrio, increased Actinomyces, and altered metabolites, including amino acids and phospholipids. Conclusions: IDA significantly impacts gut microbiota and metabolic profiles, offering insights for therapeutic strategies targeting microbiota and metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Tong Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China; (H.Z.); (Z.F.); (Y.D.); (X.L.); (C.W.); (T.Z.); (J.Z.)
| | - Jie Liang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China; (H.Z.); (Z.F.); (Y.D.); (X.L.); (C.W.); (T.Z.); (J.Z.)
| |
Collapse
|
19
|
Zhu X, Lu H, Jia H, Wei X, Xue J, Li W, Zhang J, Wang Y, Yan J, Sun H, Ge Y, Zhang Z. Ferrostatin-1 reduces the inflammatory response of rheumatoid arthritis by decreasing the antigen presenting function of fibroblast-like synoviocytes. J Transl Med 2025; 23:280. [PMID: 40050869 PMCID: PMC11884008 DOI: 10.1186/s12967-025-06300-0] [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: 11/30/2024] [Accepted: 02/23/2025] [Indexed: 03/10/2025] Open
Abstract
Rheumatoid arthritis (RA) is a systemic chronic autoimmune disease with complex mechanism. Currently, ferroptosis is believed to play a role in it, but the specific mechanism is unknown, especially in immune response. In this study, we demonstrated that the high expression of major histocompatibility complex I (MHC-I) molecules in RA fibroblast-like synoviocytes (FLSs) is an antigen-presenting cell property and that this property is closely related to the increase in antigens after citrullination. Moreover, we detected higher levels of ferroptosis among FLSs from RA patient than among FLSs from OA patients. Ferroptosis can increase the expression of citrullinated histone H3 (cit-h3) by promoting the production of peptidyl arginine deiminase 4 (PAD4), which further promotes the expression of MHC-I molecules. We cocultured RA-FLSs treated with ferroptosis drugs with selected CD8 + T cells to assess the effect of ferroptosis on the endogenous antigen-presenting function of RA-FLSs. Ferroptosis promoted the proliferation of CD8 + T cells and the release of the inflammatory factors Tumor necrosis factor-α (TNF-α) and Interferon-gamma (IFN-γ), which enhanced the inflammatory effect. This phenomenon was also observed in a collagen-induced arthritis (CIA) mouse model. Finally, ferrostatin-1 (fer-1), a ferroptosis inhibitor, inhibited the above effects and reduced the release of inflammatory factors, indicating that ferroptosis may play a therapeutic role in RA and providing new ideas for the treatment of RA in the field of immunity.
Collapse
Affiliation(s)
- Xiaoying Zhu
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hanya Lu
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haonan Jia
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuemin Wei
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawei Xue
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjing Li
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Juan Zhang
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanli Wang
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingyao Yan
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haoyuan Sun
- Department of Osteology, Heilongjiang Provincial Hospital, Harbin, China
| | - Yanlei Ge
- Department of Respiratory Medicine, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Zhiyi Zhang
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
20
|
Zhang J, Wang F, Sun Z, Ye J, Chu H. Multidimensional applications of prussian blue-based nanoparticles in cancer immunotherapy. J Nanobiotechnology 2025; 23:161. [PMID: 40033359 PMCID: PMC11874808 DOI: 10.1186/s12951-025-03236-x] [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: 10/25/2024] [Accepted: 02/16/2025] [Indexed: 03/05/2025] Open
Abstract
Immunotherapy holds notable progress in the treatment of cancer. However, the clinical therapeutic effect remains a significant challenge due to immune-related side effects, poor immunogenicity, and immunosuppressive microenvironment. Nanoparticles have emerged as a revolutionary tool to surmount these obstacles and amplify the potency of immunotherapeutic agents. Prussian blue nanoparticles (PBNPs) exhibit multi-dimensional immune function in cancer immunotherapy, including acting as a nanocarrier to deliver immunotherapeutic agents, as a photothermal agent to improve the efficacy of immunotherapy through photothermal therapy, as a nanozyme to regulate tumor microenvironment, and as an iron donor to induce immune events related to ferroptosis and tumor-associated macrophages polarization. This review focuses on the advances and applications of PBNPs in cancer immunotherapy. First, the biomedical functions of PBNPs are introduced. Then, based on the immune function of PBNPs, we systematically reviewed the multidimensional application of PBNPs in cancer immunotherapy. Finally, the challenges and future developments of PBNPs-based cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
21
|
Sun X, Zhang C, Fan B, Liu Q, Shi X, Wang S, Chen T, Cai X, Hu C, Sun H, Puno P, Cao P. Cotargeting of thioredoxin 1 and glutamate-cysteine ligase in both imatinib-sensitive and imatinib-resistant CML cells. Biochem Pharmacol 2025; 233:116763. [PMID: 39832669 DOI: 10.1016/j.bcp.2025.116763] [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: 11/04/2024] [Revised: 01/04/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Chronic myeloid leukemia (CML) is a type of malignancy characterized by harboring the oncogene Bcr-Abl, which encodes the constitutively activated tyrosine kinase BCR-ABL. Although tyrosine kinase inhibitors targeting BCR-ABL have revolutionized CML therapy, native and acquired drug resistance commonly remains a great challenge. Thioredoxin 1 (Trx1) and glutamate-cysteine ligase (GCL), which are two major antioxidants that maintain cellular redox homeostasis, are potential targets for cancer therapy and overcoming drug resistance. However, how their inhibition is implicated in CML is still unclear. Here, our results revealed that Trx1 was overexpressed in patients with CML compared with healthy donors. Trx1 expression was greater in imatinib-resistant CML cells than in imatinib-sensitive cells. Pharmacological inhibitors of Trx1 attenuated cell growth and reduced colony formation in both imatinib-sensitive and imatinib-resistant CML cells. Furthermore, decreased Trx1 expression enhanced the cytotoxicity of the GCL inhibitor buthionine sulfoximine (BSO). We surmise that the combined inhibition of Trx1 and GCL promotes the induction of hydrogen peroxide and depletes GPX4 expression in CML cells, resulting in ferroptosis in cancerous cells. Finally, the combined inhibition of Trx1 and GCL had a synergistic effect on CML cells in murine xenograft models. These findings offer crucial informationregarding the combined roles ofTrx1 and GCL in triggering ferroptosis in CML and suggestefficacioustherapeutic uses for these systems in this disease.
Collapse
MESH Headings
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Humans
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/physiology
- Thioredoxins/antagonists & inhibitors
- Thioredoxins/metabolism
- Thioredoxins/genetics
- Thioredoxins/biosynthesis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Animals
- Glutamate-Cysteine Ligase/antagonists & inhibitors
- Glutamate-Cysteine Ligase/metabolism
- Glutamate-Cysteine Ligase/genetics
- Mice
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Female
- Male
- Cell Line, Tumor
- Xenograft Model Antitumor Assays/methods
- K562 Cells
Collapse
Affiliation(s)
- Xiaoyan Sun
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Chunli Zhang
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Bo Fan
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Qingyu Liu
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Xiaofeng Shi
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Shuxia Wang
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Ting Chen
- Hematology, The People's Hospital of Rugao, Jiangsu, PR China
| | - Xueting Cai
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Chunping Hu
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Handong Sun
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, PR China
| | - Pematenzin Puno
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, PR China.
| | - Peng Cao
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, RP China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China.
| |
Collapse
|
22
|
Xia F, Zhou X, Xiong Y, Yin C, Wang M, Li L. Development and internal validation of a nomogram for predicting recurrent respiratory tract infections in children. Respir Med 2025; 238:107961. [PMID: 39855478 DOI: 10.1016/j.rmed.2025.107961] [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: 05/15/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVE This study aimed to develop and internally validate a nomogram in predicting the risk of recurrent respiratory tract infection (RRTI) in children. METHODS A retrospective analysis was performed, involving 150 children with RRTI and 151 healthy controls, aged 0-14 years, admitted to or selected from the Pediatric Department of Yixing Hospital of Traditional Chinese Medicine between June 2022 and June 2023. Data were gathered through a comprehensive questionnaire survey on risk factors associated with RRTI. The dataset was randomly divided into a training cohort (n = 211) and a validation cohort (n = 90) in a 7:3 ratio. Significant variables were selected using LASSO regression in the training cohort to construct the nomogram, the performance of which was evaluated through Receiver Operating Characteristic (ROC) curves, calibration plots, and Decision Curve Analysis (DCA). RESULTS The LASSO regression identified five predictors in the training cohort: picky eating, age at first antibiotic use, antibiotic use within the previous year, allergic conditions, secondhand smoke exposure. Based on them, the nomogram exhibited an excellent discriminative ability, with an AUC of 0.902 (95 % CI: 0.860-0.944) and a C-index of 0.902 in the training cohort. The validation cohort showed an AUC of 0.826 (95 % CI: 0.742-0.909) and a C-index of 0.826, confirming a high predictive accuracy. Calibration plots showed close alignment with the ideal reference line, and DCA indicated a significant clinical net benefit. CONCLUSION Our nomogram can efficiently predict RRTI risk in children, thereby providing a personalized and graphical tool for early identification and intervention.
Collapse
Affiliation(s)
- Fei Xia
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital, Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, China; Department of Pediatrics, Yixing Hospital of Traditional Chinese Medicine, Yixing, 214200, China
| | - Xi Zhou
- Department of Pediatrics, Yixing Hospital of Traditional Chinese Medicine, Yixing, 214200, China
| | - Yan Xiong
- Department of Pediatrics, Yixing Hospital of Traditional Chinese Medicine, Yixing, 214200, China
| | - Chenghui Yin
- Department of Pediatrics, Yixing Hospital of Traditional Chinese Medicine, Yixing, 214200, China
| | - Minhua Wang
- Department of Pediatrics, Yixing Hospital of Traditional Chinese Medicine, Yixing, 214200, China.
| | - Ling Li
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital, Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, China; Department of Respiratory Medicine & Clinical Allergy Center, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, 214023, China.
| |
Collapse
|
23
|
Pei Z, Fan J, Tang M, Li Y. Ferroptosis: A New Strategy for the Treatment of Fibrotic Diseases. Adv Biol (Weinh) 2025; 9:e2400383. [PMID: 39377183 DOI: 10.1002/adbi.202400383] [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: 07/02/2024] [Revised: 09/07/2024] [Indexed: 10/09/2024]
Abstract
Ferroptosis is a new type of cell death characterized by iron dependence and the excessive accumulation of lipid reactive oxygen species (lipid ROS) that has gradually become better characterized. There is sufficient evidence indicating that ferroptosis is associated with a variety of human life activities and diseases, such as tumor suppression, ischemic organ injury, and degenerative disorders. Notably, ferroptosis is also involved in the initiation and development of fibrosis in various organs, including liver fibrosis, pulmonary fibrosis, renal fibrosis, and cardiac fibrosis, which is usually irreversible and refractory. Although a large number of patients with fibrosis urgently need to be treated, the current treatment options are still limited and unsatisfactory. Organ fibrosis involves a series of complex and orderly processes, such as parenchymal cell damage, recruitment of inflammatory cells and activation of fibroblasts, which ultimately leads to the accumulation of extracellular matrix (ECM) and the formation of fibrosis. An increasing number of studies have confirmed the close association between these pathological processes and ferroptosis. This review summarizes the role and function of ferroptosis in fibrosis and proposes several potential therapeutic strategies and pathways based on ferroptosis.
Collapse
Affiliation(s)
- Zhuo Pei
- Air Force Hospital of the Central Theater Command of PLA, Datong, 037006, China
| | - Jing Fan
- Air Force Hospital of the Northern Theater Command of the People's Liberation Army of China, Shenyang, 110044, China
| | - Maolin Tang
- Air Force Hospital of the Central Theater Command of PLA, Datong, 037006, China
| | - Yuhong Li
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| |
Collapse
|
24
|
Bu X, Wang L. Iron metabolism and the tumor microenvironment: A new perspective on cancer intervention and therapy (Review). Int J Mol Med 2025; 55:39. [PMID: 39749705 PMCID: PMC11722052 DOI: 10.3892/ijmm.2024.5480] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/29/2024] [Indexed: 01/04/2025] Open
Abstract
Iron metabolism plays a crucial role in the tumor microenvironment, influencing various aspects of cancer cell biology and tumor progression. This review discusses the regulatory mechanisms of iron metabolism within the tumor microenvironment and highlights how tumor cells and associated stromal cells manage iron uptake, accumulation and regulation. The sources of iron within tumors and the biological importance of ferroptosis in cancer were explored, focusing on its mechanisms, biological effects and, in particular, its tumor‑suppressive properties. Furthermore, the protective strategies employed by cancer cells to evade ferroptosis were examined. This review also delves into the intricate relationship between iron metabolism and immune modulation within the tumor microenvironment, detailing the impact on tumor‑associated immune cells and immune evasion. The interplay between ferroptosis and immunotherapy is discussed and potential strategies to enhance cancer immunotherapy by modulating iron metabolism are presented. Finally, the current ferroptosis‑based cancer therapeutic approaches were summarized and future directions for therapies that target iron metabolism were proposed.
Collapse
Affiliation(s)
- Xiaorui Bu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lufang Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
25
|
Li Y, Huang B, Yuan M, Zhang C, Zhang X, Hao J, Tao F, Geng F, Wang G, Su P. Associations between serum metal mixtures and systemic inflammation indices among Chinese early adolescents: A prospective cohort study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117952. [PMID: 40014987 DOI: 10.1016/j.ecoenv.2025.117952] [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: 12/17/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Research has demonstrated a link between metal exposure and inflammation. However, little is known about this relationship among adolescents, especially in prospective cohort studies. The aim of this study was to investigate the relationship between serum metal exposure and inflammatory status in Chinese early adolescents. METHODS In this study, 12 serum metals were detected at baseline in 1551 participants from the Chinese Early Adolescents Cohort. The participants' inflammatory status was assessed via three systemic inflammation indices (neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII)) at both baseline and follow-up. Generalized linear mixed models and restricted cubic splines regression were used to examine the linear and nonlinear relationships between single metal concentrations and systemic inflammation indices. Multiple mixture models were implemented to assess the relationships of mixed metals with systemic inflammation indices. Additionally, sex subgroup analyses were conducted to explore the sex-specific associations between serum metals and inflammatory status. RESULTS Single-exposure analysis revealed that exposure to multiple serum metals, such as chromium, cobalt, copper and lead, was positively associated with the NLR and SII, whereas iron was negatively correlated with the three systemic inflammation indices (PFDR<0.05). Additionally, inverted U-shaped associations were observed between vanadium, manganese and systemic inflammation indices. According to the mixture models, high levels of the serum metal mixture were positively correlated with the NLR and the SII. Cobalt had the highest positive weight in the mixed samples, whereas iron had the greatest negative weight in the serum-metal mixtures. Subgroup analyses revealed that serum exposure to the metal mixture had a more significant effect on systemic inflammation markers in females than in males. CONCLUSIONS This study reveals the impact of real-world mixed metal exposure on adolescents' inflammatory levels, which is of primary significance for protecting the healthy development of early adolescents.
Collapse
Affiliation(s)
- Yonghan Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Binbin Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui 230032, China
| | - Mengyuan Yuan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Chao Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Xueying Zhang
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jiahu Hao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui 230032, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui 230032, China
| | - Feng Geng
- Department of Psychology and Sleep Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China
| | - Gengfu Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui 230032, China.
| | - Puyu Su
- Department of Psychology and Sleep Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China; School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
26
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [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: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
27
|
Feng W, Weng Y, Shi W, Liang S, Liao X, Chu R, Ai Q, Mai K, Wan M. Aquatic high iron induces hepatic ferroptosis in zebrafish (Danio rerio) via interleukin-22 signaling pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125627. [PMID: 39746632 DOI: 10.1016/j.envpol.2024.125627] [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: 08/13/2024] [Revised: 12/26/2024] [Accepted: 12/31/2024] [Indexed: 01/04/2025]
Abstract
Iron is one of the indispensable trace elements in living organisms. However, excessive iron deposition in organisms is prone to induce dysfunction of the liver and other vital organs. The present study aimed to investigate the mechanism how aquatic high iron affects iron transport and induces hepatic injury in zebrafish. Our results showed that the iron levels in zebrafish liver and serum were significantly increased after the fish treated with aquatic high iron (200 mg/L ferric ammonium citrate, FAC) for 21 days. Meanwhile, hepatic fibrosis was observed in zebrafish with high iron treatment. Furthermore, the expression of hepcidin, a key factor in the regulation of iron homeostasis, as well as other factors related to iron transport, was significantly influenced by high iron treatment. Nonetheless, different tissues, such as liver, gill and gut, diversely responded to high iron in water. Interestingly, our results identified that the expression of IL-22, instead of IL-6, was significantly elevated after high iron treatment. Moreover, high iron triggered STAT3 phosphorylation via IL-22, leading to the augmented expression of hepcidin and hepatic iron accumulation. As a result, the iron overload in fish liver induced hepatic ferroptosis, marked as the repressed activity of glutathione peroxidase (GPx) and elevated lipid peroxidation. Further studies confirmed that, unlike wild-type (WT) zebrafish, the expression of hepcidin and iron content in the liver of il22-deficient zebrafish was unaffected upon to high iron treatment. At the meantime, hepatic ferroptosis and fibrosis induced by high iron was significantly alleviated in il22-deficient zebrafish. In summary, aquatic high iron induced hepcidin expression in zebrafish by activating the IL-22/STAT3 signaling pathway, which in turn regulated hepatic iron transport and ferroptosis in zebrafish. The present study identified for the first time that IL-22 may be a potential regulatory target for iron overload-induced liver injury.
Collapse
Affiliation(s)
- Wei Feng
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Yizhuo Weng
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Wenkai Shi
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Shufei Liang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Xinmeng Liao
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Ruixia Chu
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong, 266237, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong, 266237, China
| | - Min Wan
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong, 266237, China.
| |
Collapse
|
28
|
Han H, Zhang G, Yang Y, Li C, Li X, Zhong L, Chen Z, Xiong J, Cai T, Zhang L, Zhang X, Zhao Q. Therapeutic potential of monomethyl fumarate and aluminum ion combination in alleviating inflammation and oxidative stress in psoriasis. Redox Biol 2025; 79:103482. [PMID: 39736200 PMCID: PMC11750270 DOI: 10.1016/j.redox.2024.103482] [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: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/22/2024] [Indexed: 01/01/2025] Open
Abstract
Psoriasis is a chronic inflammatory skin condition characterized by erythematous plaques with white scales. Its pathogenesis is closely linked to oxidative stress and an imbalance in Th1/Th2 immune responses. Current treatments for psoriasis, such as topical agents, systemic therapies and phototherapy, frequently fail to achieve complete remission in clinical settings. Monomethyl fumarate (MMF), which has been approved by the US Food and Drug Administration in 2020 for multiple sclerosis, has demonstrated efficacy in psoriasis management. Additionally, our previous studies have identified aluminum ions as beneficial in psoriasis treatment. This present study investigates the combined therapeutic effects of MMF and aluminum ions and observed that the combination treatment achieves superior efficacy compared to either treatment alone in a psoriasis mouse model through the modulation of the Nrf2/NF-κB signaling pathway, as demonstrated in cellular models. The combination first activates Nrf2 nuclear translocation and induces antioxidant gene expression, followed by the inhibition of NF-κB nuclear translocation and phosphorylation, which reduces Th1 cytokine production and cellular chemotaxis. Concurrently, the treatment elevates Th2 cytokine secretion, thereby increasing the anti-inflammatory response in HaCaT cells. Overall, these findings support the MMF and aluminum ions combination (MMFAL) as a potential therapeutic strategy for psoriasis, effectively diminishing inflammation and oxidative stress.
Collapse
Affiliation(s)
- Hang Han
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Guojiang Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yuanyuan Yang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Chenxi Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xiandeng Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Ling Zhong
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Zan Chen
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jianxia Xiong
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Cai
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingjuan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Xiao Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
29
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2025; 480:759-784. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [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: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
30
|
Hanna M, Akabawy AMA, Khalifa MM, Elbaset MA, Imam RA, Seddiek H. Intracellular iron accumulation throughout the progression of sepsis influences the phenotype and function of activated macrophages in renal tissue damage. Front Physiol 2025; 16:1430946. [PMID: 39949667 PMCID: PMC11821637 DOI: 10.3389/fphys.2025.1430946] [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: 05/10/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
Sepsis, the most common cause of acute kidney injury, remains a major socioeconomic burden. A dysregulated immune response leads to progressive organ dysfunction. Although numerous inflammatory pathways were described, most are still vague and need to be studied in terms of the mechanisms to improve the therapeutic intervention. We tackled the relationship between intracellular iron overload and macrophage polarization within 6, 24, and 72 h of sepsis induction. In our study, sepsis-induced kidney injury was caused by using the cecal ligation and puncture (CLP) model. Our results indicated severe renal tissue damage with a progressive increase in serum BUN and creatinine with architectural tissue damage and positive PAS staining. There was increased expression of CD8+ CD68+ M1 macrophage markers with upregulation of iNOS and co-expression of CD163+. Alternatively, Arg1+ Fizz1+ M2 macrophage markers were downregulated with increased iNOS/Arg1 ratio. TFR1, cubilin, and DMT1, as iron transport systems, were increased compared to sham but were significant after 72 h, while ZIP8 showed no significant change. There was a correlation between iron overload and M1 macrophage polarization with CD163+ phenotype, together with fibrotic changes. The intracellular iron overload with downregulation of ferritin was strongly related to macrophage polarization that was exaggerated at 72 h. Finally, early introduced therapy to target free iron during sepsis is a proposed novel solution for protecting the renal tissue from acute injury due to macrophage activation that may end up with chronic kidney injury, if not mortality.
Collapse
Affiliation(s)
- Mira Hanna
- Department of Medical Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Ahmed M. A. Akabawy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed Mansour Khalifa
- Department of Medical Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
- Department of Medical Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Marawan Abd Elbaset
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Reda Abdelnasser Imam
- Department of Anatomy and Embryology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Hanan Seddiek
- Department of Medical Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| |
Collapse
|
31
|
Tian W, Ma X, Liu H, Wang Z, Liu C, Xie C. Maternal Ferrous Sucrose Supplementation Improves Reproductive Performance of Sows and Hepatic Iron Stores of Neonatal Piglets. Animals (Basel) 2025; 15:343. [PMID: 39943113 PMCID: PMC11815870 DOI: 10.3390/ani15030343] [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: 11/29/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
As one of the most important trace elements required by sows, especially in the late gestation period, iron plays a crucial role in the growth and development of a fetus. To explore the effects of dietary supplementation with ferrous sucrose on the reproductive performance of sows and the hepatic iron stores in offspring, sixty primiparous Landrace × Yorkshire sows on day 95 of gestation with an average body weight of 174.1 ± 7.7 kg were randomly assigned to two groups of a basic diet (control) and a basic diet supplemented with 109 mg/kg ferrous sucrose (FS) in a fully randomized block design. The trial lasted for 20 days. The results showed that maternal supplementation with ferrous sucrose significantly increased litter weight (p = 0.002) in neonatal piglets. Compared with the control group, the serum iron and serum transferrin saturation of farrowing sows increased by 45.67% (p = 0.002) and 37.01% (p = 0.033), respectively, and umbilical cord serum iron (p = 0.012) also increased in the FS group. Finally, the serum iron (p < 0.001) and hepatic iron stores (p = 0.071) of neonatal piglets were both increased to varying degrees. Taken together, supplementation of pregnant sows with ferrous sucrose has positive effects on the growth and hepatic iron stores of their offspring.
Collapse
Affiliation(s)
- Wen Tian
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (W.T.)
| | - Xiaofan Ma
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (W.T.)
| | - Hongwei Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (W.T.)
| | - Zhefeng Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (W.T.)
| | - Chunxue Liu
- Anyou Biotechnology Group Co., Ltd., Taicang 215437, China
| | - Chunyan Xie
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (W.T.)
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Livestock and Poultry Health Breeding Technology Engineering Center, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China
| |
Collapse
|
32
|
Wang Y, Li C, Ba T, Wang S, He L, Chen Z, Pu J, Cui X, Jia G. Gastrointestinal dysfunction as the main performance of the oral toxicity of titanium dioxide nanoparticle on gastric ulcer rats. NANOIMPACT 2025; 37:100551. [PMID: 39986608 DOI: 10.1016/j.impact.2025.100551] [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: 11/11/2024] [Revised: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) have promising applications in food additives and pharmaceutical dressings, raising concerns about their oral safety. The current studies mainly focus on healthy groups, and the effect of TiO2 NPs on the patient population is rarely known. Here, a comprehensive toxicity study of TiO2 NPs (75 ± 15 nm, anatase) in gastric ulcer rats (male 8-week old Sprague-Dawley rats) is reported following oral exposure at dose of 0, 10, 50, 200 mg/kg body weight per day for 30 days. The gastric ulcer rats were produced by submucosal injection of acetic acid solution into the rat stomach. The healthy rats were used as the normal control. We evaluated nanoparticle biodistribution, systemic toxicity, and gastrointestinal function indices in the rats. Our findings indicate that oral administration of TiO2 NPs resulted in minimal intestinal absorption and transport with limited systemic organ toxicity. The internalization of TiO2 NPs and activation of mast cells in the stomach tissues, along with the low serum levels of histamine and IgE, suggest a localized allergic reaction rather than a systemic one. Furthermore, the notably reduced plasma levels of D-lactate and the activity of diamine oxidase (DAO) indicated the upregulation of intestinal barrier function. These statistically significant results indicated that gastrointestinal dysfunction was the main performance of the oral toxicity of TiO2 NPs on gastric ulcer rats, emphasizing the importance of controlling the intake of TiO2 NPs in patients with gastric ulcers.
Collapse
Affiliation(s)
- Yun Wang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University and Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, PR China..
| | - Chen Li
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University and Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, PR China
| | - Te Ba
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University and Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, PR China
| | - Shengyuan Wang
- Department of Occupational Health, Public Health College, Harbin Medical University, Harbin, PR China
| | - Langzhi He
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University and Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, PR China
| | - Zhangjian Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University and Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, PR China
| | - Ji Pu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University and Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, PR China
| | - Xiaoxing Cui
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University and Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, PR China
| | - Guang Jia
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University and Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, PR China..
| |
Collapse
|
33
|
Wang B, Wang J, Liu C, Li C, Meng T, Chen J, Liu Q, He W, Liu Z, Zhou Y. Ferroptosis: Latest evidence and perspectives on plant-derived natural active compounds mitigating doxorubicin-induced cardiotoxicity. J Appl Toxicol 2025; 45:135-158. [PMID: 39030835 DOI: 10.1002/jat.4670] [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: 06/08/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/22/2024]
Abstract
Doxorubicin (DOX) is a chemotherapy drug widely used in clinical settings, acting as a first-line treatment for various malignant tumors. However, its use is greatly limited by the cardiotoxicity it induces, including doxorubicin-induced cardiomyopathy (DIC). The mechanisms behind DIC are not fully understood, but its potential biological mechanisms are thought to include oxidative stress, inflammation, energy metabolism disorders, mitochondrial damage, autophagy, apoptosis, and ferroptosis. Recent studies have shown that cardiac injury induced by DOX is closely related to ferroptosis. Due to their high efficacy, availability, and low side effects, natural medicine treatments hold strong clinical potential. Currently, natural medicines have been shown to mitigate DOX-induced ferroptosis and ease DIC through various functions such as antioxidation, iron ion homeostasis correction, lipid metabolism regulation, and mitochondrial function improvement. Therefore, this review summarizes the mechanisms of ferroptosis in DIC and the regulation by natural plant products, with the expectation of providing a reference for future research and development of inhibitors targeting ferroptosis in DIC. This review explores the mechanisms of ferroptosis in doxorubicin-induced cardiomyopathy (DIC) and summarizes how natural plant products can alleviate DIC by inhibiting ferroptosis through reducing oxidative stress, correcting iron ion homeostasis, regulating lipid metabolism, and improving mitochondrial function.
Collapse
Affiliation(s)
- Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiameng Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wang He
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiping Liu
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
34
|
Wu T, Ji M, Li T, Luo L. The molecular and metabolic landscape of ferroptosis in respiratory diseases: Pharmacological aspects. J Pharm Anal 2025; 15:101050. [PMID: 40034685 PMCID: PMC11873008 DOI: 10.1016/j.jpha.2024.101050] [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: 02/29/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 03/05/2025] Open
Abstract
Ferroptosis is a form of cell death that occurs when there is an excess of reactive oxygen species (ROS), lipid peroxidation, and iron accumulation. The precise regulation of metabolic pathways, including iron, lipid, and amino acid metabolism, is crucial for cell survival. This type of cell death, which is associated with oxidative stress, is controlled by a complex network of signaling molecules and pathways. It is also implicated in various respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), lung cancer, pulmonary fibrosis (PF), and the coronavirus disease 2019 (COVID-19). To combat drug resistance, it is important to identify appropriate biological markers and treatment targets, as well as intervene in respiratory disorders to either induce or prevent ferroptosis. The focus is on the role of ferroptosis in the development of respiratory diseases and the potential of targeting ferroptosis for prevention and treatment. The review also explores the interaction between immune cell ferroptosis and inflammatory mediators in respiratory diseases, aiming to provide more effective strategies for managing cellular ferroptosis and respiratory disorders.
Collapse
Affiliation(s)
- Tong Wu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Miaorong Ji
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
35
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
36
|
Zhang Y, Li H, Chen Y, Li C, Ye H, Qiu J, Liu X, Sun W, Zhang X, Tian N, Zhou Y. Nordihydroguaiaretic acid suppresses ferroptosis and mitigates intervertebral disc degeneration through the NRF2/GPX4 axis. Int Immunopharmacol 2024; 143:113590. [PMID: 39541847 DOI: 10.1016/j.intimp.2024.113590] [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: 05/20/2024] [Revised: 10/13/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Intervertebral disc degeneration (IVDD) is a major contributor to low back pain (LBP), while LBP is the leading cause of disability. However, the effective pharmacological interventions for IVDD are still lacking. Studies have elucidated that ferroptosis plays a crucial role in the pathogenesis of IVDD. This study aimed to evaluate the effects of various natural products, specifically screening for those that suppress ferroptosis induced in nucleus pulposus cells (NPCs) via RSL3. Previously, we have identified that a list of natural products in the library may suppress oxidative stress damage in NPCs, while oxidative stress is a major contributor to ferroptosis. The current study sought to verify the ferroptosis inhibitory effect of these products in NPCs. Through screening of the top 20 natural products in the list, we found that Nordihydroguaiaretic acid (NDGA) was the most effective compound to inhibit ferroptosis in NPCs. Mechanism study demonstrated that NDGA may promote the nuclear expression of the key transcriptional factor nuclear factor erythroid 2-related factor 2 (Nrf2), which subsequently increase the expression of the ferroptosis suppressor gene GPX4, and reduce the degradation of the extracellular matrix (ECM) and suppress the progression of inflammation. In the rat puncture induced IVDD model, intraperitoneal injection of NDGA delayed the progression of IVDD. In conclusion, our study indicates that NDGA is a potential drug for the treatment of IVDD.
Collapse
Affiliation(s)
- Yekai Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China
| | - Hualin Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China
| | - Yiji Chen
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China; School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Chenchao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China
| | - Haobo Ye
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China
| | - Jiawei Qiu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China
| | - Xiaopeng Liu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China
| | - Weiqian Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China; Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, 310000 Zhejiang Province, China.
| | - Naifeng Tian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China; Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, 310000 Zhejiang Province, China.
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325088 Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325035 Zhejiang Province, China; Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, 310000 Zhejiang Province, China.
| |
Collapse
|
37
|
Zheng Y, Yan F, He S, Luo L. Targeting ferroptosis in autoimmune diseases: Mechanisms and therapeutic prospects. Autoimmun Rev 2024; 23:103640. [PMID: 39278299 DOI: 10.1016/j.autrev.2024.103640] [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: 06/25/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Ferroptosis is a form of regulated cell death that relies on iron and exhibits unique characteristics, including disrupted iron balance, reduced antioxidant defenses, and abnormal lipid peroxidation. Recent research suggests that ferroptosis is associated with the onset and progression of autoimmune disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and multiple sclerosis (MS). However, the precise effects and molecular mechanisms remain incompletely understood. This article presents an overview of how ferroptosis mechanisms contribute to the development and advancement of autoimmune diseases, as well as the involvement of various immune cells in linking ferroptosis to autoimmune conditions. It also explores potential drug targets within the ferroptosis pathway and recent advancements in therapeutic approaches aimed at preventing and treating autoimmune diseases by targeting ferroptosis. Lastly, the article discusses the challenges and opportunities in utilizing ferroptosis as a potential therapeutic avenue for autoimmune disorders.
Collapse
Affiliation(s)
- Yingzi Zheng
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Fangfang Yan
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China.
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
38
|
Liu J, Feng G. The causal relationship between trace element status and upper gastrointestinal ulcers: a Mendelian randomization study. Front Nutr 2024; 11:1443090. [PMID: 39539362 PMCID: PMC11557352 DOI: 10.3389/fnut.2024.1443090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background This study aimed to investigate the bidirectional causal relationships between trace elements (such as zinc, magnesium, phosphate, and folate) and upper gastrointestinal ulcers (including gastric and duodenal ulcers). We utilized a two-sample Mendelian randomization (MR) analysis to achieve this. Methods We conducted a two-sample MR analysis using summary-level data from genome-wide association studies (GWAS) obtained from public genomics repositories. We utilized a range of MR methods, including inverse-variance weighted (IVW), MR-Egger, and weighted median methods, and conducted a meta-analysis to synthesize results across different datasets. To ensure the robustness of our findings, we performed extensive sensitivity analyses, including pleiotropy assessment, heterogeneity tests, and leave-one-out analysis. Results Our findings are significant, indicating a positive causal relationship between increased zinc levels and the risk of gastric ulcers. Moreover, magnesium and folate appear to offer potential protective effects against gastroduodenal ulcers (p < 0.05). The meta-analysis further supports the causal relationship between zinc and gastric ulcers (p < 0.05), confirming zinc's significant causal impact on this condition. Conclusion The study confirms a positive causal relationship between zinc and gastric ulcers and highlights the complexity of how trace elements regulate the progression of upper gastrointestinal ulcers. These results provide a scientific basis for dietary recommendations regarding trace element intake in clinical and public health practices. They also offer new insights into effective prevention and treatment strategies for gastric and duodenal ulcers.
Collapse
Affiliation(s)
- Jianwei Liu
- Department of Gastroenterology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Gege Feng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
39
|
Belbellaj W, Lona-Durazo F, Bodano C, Busseuil D, Cyr MC, Fiorillo E, Mulas A, Provost S, Steri M, Tanaka T, Vanderwerff B, Wang J, Byrne RP, Cucca F, Dubé MP, Ferrucci L, McLaughlin RL, Tardif JC, Zawistowski M, Gagliano Taliun SA. The role of genetically predicted serum iron levels on neurodegenerative and cardiovascular traits. Sci Rep 2024; 14:24588. [PMID: 39427026 PMCID: PMC11490554 DOI: 10.1038/s41598-024-76245-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
Iron is an essential mineral that supports numerous biological functions. Studies have reported associations between iron dysregulation and certain cardiovascular and neurodegenerative diseases, but the direction of influence is not clear. Our goal was to use computational approaches to better understand the role of genetically predicted iron levels on disease risk. We meta-analyzed genome-wide association study summary statistics for serum iron levels from two cohorts and two previous meta-analyses. We then obtained summary statistics from 11 neurodegenerative, cerebrovascular, cardiovascular or lipid traits to assess global and regional genetic correlation between iron levels and these traits. We used two-sample Mendelian randomization (MR) to estimate causal effects. Sex-stratified analyses were also carried out to identify effects potentially differing by sex. Overall, we identified three significant global correlations between iron levels and (i) coronary heart disease, (ii) triglycerides, and (iii) high-density lipoprotein (HDL) cholesterol levels. A total of 194 genomic regions had significant (after correction for multiple testing) local correlations between iron levels and the 11 tested traits. MR analysis revealed two potential causal relationships, between genetically predicted iron levels and (i) total cholesterol or (ii) non-HDL cholesterol. Sex-stratified analyses suggested a potential protective effect of iron levels on Parkinson's disease risk in females, but not in males. Our results will contribute to a better understanding of the genetic basis underlying iron in cardiovascular and neurological health in aging, and to the eventual identification of new preventive interventions or therapeutic avenues for diseases which affect women and men worldwide.
Collapse
Affiliation(s)
- Wiame Belbellaj
- Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
| | - Frida Lona-Durazo
- Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
| | - Cinzia Bodano
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 09042, Monserrato-Cagliari, Italy
| | - David Busseuil
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
| | - Marie-Christyne Cyr
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, H1T 1C8, Canada
| | - Edoardo Fiorillo
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 08045, Lanusei, Italy
| | - Antonella Mulas
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 08045, Lanusei, Italy
| | - Sylvie Provost
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, H1T 1C8, Canada
| | - Maristella Steri
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 09042, Monserrato-Cagliari, Italy
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institutes on Aging, Baltimore, MD, USA
| | - Brett Vanderwerff
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jiongming Wang
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ross P Byrne
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, D02 DK07, Republic of Ireland
| | - Francesco Cucca
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Marie-Pierre Dubé
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, H1T 1C8, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institutes on Aging, Baltimore, MD, USA
| | - Russell L McLaughlin
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, D02 DK07, Republic of Ireland
| | - Jean-Claude Tardif
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Matthew Zawistowski
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sarah A Gagliano Taliun
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
| |
Collapse
|
40
|
Lan W, Yang L, Tan X. Crosstalk between ferroptosis and macrophages: potential value for targeted treatment in diseases. Mol Cell Biochem 2024; 479:2523-2543. [PMID: 37880443 DOI: 10.1007/s11010-023-04871-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023]
Abstract
Ferroptosis is a newly identified form of programmed cell death that is connected to iron-dependent lipid peroxidization. It involves a variety of physiological processes involving iron metabolism, lipid metabolism, oxidative stress, and biosynthesis of nicotinamide adenine dinucleotide phosphate, glutathione, and coenzyme Q10. So far, it has been discovered to contribute to the pathological process of many diseases, such as myocardial infarction, acute kidney injury, atherosclerosis, and so on. Macrophages are innate immune system cells that regulate metabolism, phagocytize pathogens and dead cells, mediate inflammatory reactions, promote tissue repair, etc. Emerging evidence shows strong associations between macrophages and ferroptosis, which can provide us with a deeper comprehension of the pathological process of diseases and new targets for the treatments. In this review, we summarized the crosstalk between macrophages and ferroptosis and anatomized the application of this association in disease treatments, both non-neoplastic and neoplastic diseases. In addition, we have also addressed problems that remain to be investigated, in the hope of inspiring novel therapeutic strategies for diseases.
Collapse
Affiliation(s)
- Wanxin Lan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Xuelian Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
41
|
Wang J, Lv C, Wei X, Li F. Molecular mechanisms and therapeutic strategies for ferroptosis and cuproptosis in ischemic stroke. Brain Behav Immun Health 2024; 40:100837. [PMID: 39228970 PMCID: PMC11369453 DOI: 10.1016/j.bbih.2024.100837] [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: 01/15/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 09/05/2024] Open
Abstract
Ischemic stroke, as one of the most severe and prevalent neurological disorders, poses a significant threat to the health and quality of life of affected individuals. Stemming from the obstruction of blood flow, ischemic stroke, leads to cerebral tissue hypoxia and ischemia, instigating a cascade of pathophysiological changes that markedly exacerbate neuronal damage and may even culminate in cell death. In recent years, emerging research has increasingly focused on novel cell death mechanisms such as ferroptosis and cuproptosis. Mounting evidence underscores the independent roles of ferroptosis and cuproptosis in ischemic stroke. This review aims to elucidate potential cross-regulatory mechanisms between ferroptosis and cuproptosis, exploring their regulatory roles in ischemic stroke. The objective is to provide targeted therapeutic intervention strategies.
Collapse
Affiliation(s)
- Jing Wang
- Department of neurology, Lu 'an Municipal People's Hospital, Anhui, China
- Bengbu Medical College, Anhui, China
| | - Cunming Lv
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Xinyu Wei
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Feng Li
- Department of neurology, Lu 'an Municipal People's Hospital, Anhui, China
| |
Collapse
|
42
|
An W, Li T, Tian X, Fu X, Li C, Wang Z, Wang J, Wang X. Allergies to Allergens from Cats and Dogs: A Review and Update on Sources, Pathogenesis, and Strategies. Int J Mol Sci 2024; 25:10520. [PMID: 39408849 PMCID: PMC11476515 DOI: 10.3390/ijms251910520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Inhalation allergies caused by cats and dogs can lead to a range of discomforting symptoms, such as rhinitis and asthma, in humans. With the increasing popularity of and care provided to these companion animals, the allergens they produce pose a growing threat to susceptible patients' health. Allergens from cats and dogs have emerged as significant risk factors for triggering asthma and allergic rhinitis worldwide; however, there remains a lack of systematic measures aimed at assisting individuals in recognizing and preventing allergies caused by these animals. This review provides comprehensive insights into the classification of cat and dog allergens, along with their pathogenic mechanisms. This study also discusses implementation strategies for prevention and control measures, including physical methods, gene-editing technology, and immunological approaches, as well as potential strategies for enhancing allergen immunotherapy combined with immunoinformatics. Finally, it presents future prospects for the prevention and treatment of human allergies caused by cats and dogs. This review will improve knowledge regarding allergies to cats and dogs while providing insights into potential targets for the development of next-generation treatments.
Collapse
Affiliation(s)
- Wei An
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, No. 20, Dongda Street, Beijing 100071, China;
| | - Xinya Tian
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiaoxin Fu
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Chunxiao Li
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zhenlong Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Jinquan Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiumin Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
43
|
Deng Z, Li D, Wang L, Lan J, Wang J, Ma Y. Activation of GABA BR Attenuates Intestinal Inflammation by Reducing Oxidative Stress through Modulating the TLR4/MyD88/NLRP3 Pathway and Gut Microbiota Abundance. Antioxidants (Basel) 2024; 13:1141. [PMID: 39334800 PMCID: PMC11428452 DOI: 10.3390/antiox13091141] [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: 06/10/2024] [Revised: 06/28/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Oxidative stress emerges as a prominent factor in the onset and progression of intestinal inflammation, primarily due to its critical role in damaging cells and tissues. GABAergic signaling is important in the occurrence and development of various intestinal disorders, yet its effect on oxidative stress remains unclear. We attempted to assess whether GABAergic signaling participated in the regulation of oxidative stress during enteritis. The results showed that lipopolysaccharide (LPS) significantly decreased γ-aminobutyric acid (GABA) levels in the ileal tissues of mice. Interestingly, the application of GABA significantly repressed the shedding of intestinal mucosal epithelial cells and inflammatory cell infiltration, inhibited the expressions of proinflammatory factors, including granulocyte colony-stimulating factor and granulocyte-macrophage colony stimulating factor, and enhanced the levels of anti-inflammatory cytokines interleukin (IL)-4 and IL-10, indicating that GABA could alleviate enteritis in mice. This observation was further supported by transcriptome sequencing, revealing a total of 271 differentially expressed genes, which exhibited a marked enrichment of inflammatory and immune-related pathways, alongside a prominent enhancement of GABA B receptor (GABABR) signaling following GABA administration. Effectively, Baclofen pretreatment alleviated intestinal mucosal damage in LPS-induced mice, suppressed proinflammatory cytokines IL-1β, IL-6, and tumor necrosis factor alpha expressions, and boosted total antioxidant capacity, superoxide dismutase (SOD), and glutathione (GSH) levels. Moreover, Baclofen notably enhanced the viability of LPS-stimulated IPEC-J2 cells, contracted the proinflammatory secretion factors, and reinforced SOD, GSH, and catalase levels, emphasizing the anti-inflammatory and antioxidant effects associated with GABABR activation. Mechanistically, Baclofen restrained the mRNA and protein levels of toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing 3 (NLRP3), and inducible nitric oxide synthase, while elevating nuclear factor erythroid 2-related factor 2 and heme oxygenase-1 in both mice and IPEC-J2 cells, indicating that activating GABABR strengthened antioxidant abilities by interrupting the TLR4/MyD88/NLRP3 pathway. Furthermore, 16S rDNA analysis demonstrated that Baclofen increased the relative abundance of probiotic, particularly Lactobacillus, renowned for its antioxidant properties, while reducing the relative richness of harmful bacteria, predominantly Enterobacteriaceae, suggesting that GABABR signaling may have contributed to reversing intestinal flora imbalances to relieve oxidative stress in LPS-induced mice. Our study identified previously unappreciated roles for GABABR signaling in constricting oxidative stress to attenuate enteritis, thus offering novel insights for the treatment of intestinal inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunfei Ma
- State Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.D.); (D.L.); (L.W.); (J.L.); (J.W.)
| |
Collapse
|
44
|
Carolin A, Frazer D, Yan K, Bishop CR, Tang B, Nguyen W, Helman SL, Horvat J, Larcher T, Rawle DJ, Suhrbier A. The effects of iron deficient and high iron diets on SARS-CoV-2 lung infection and disease. Front Microbiol 2024; 15:1441495. [PMID: 39296289 PMCID: PMC11408339 DOI: 10.3389/fmicb.2024.1441495] [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: 05/31/2024] [Accepted: 08/22/2024] [Indexed: 09/21/2024] Open
Abstract
Introduction The severity of Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is often dictated by a range of comorbidities. A considerable literature suggests iron deficiency and iron overload may contribute to increased infection, inflammation and disease severity, although direct causal relationships have been difficult to establish. Methods Here we generate iron deficient and iron loaded C57BL/6 J mice by feeding standard low and high iron diets, with mice on a normal iron diet representing controls. All mice were infected with a primary SARS-CoV-2 omicron XBB isolate and lung inflammatory responses were analyzed by histology, immunohistochemistry and RNA-Seq. Results Compared with controls, iron deficient mice showed no significant changes in lung viral loads or histopathology, whereas, iron loaded mice showed slightly, but significantly, reduced lung viral loads and histopathology. Transcriptional changes were modest, but illustrated widespread dysregulation of inflammation signatures for both iron deficient vs. controls, and iron loaded vs. controls. Some of these changes could be associated with detrimental outcomes, whereas others would be viewed as beneficial. Discussion Diet-associated iron deficiency or overload thus induced modest modulations of inflammatory signatures, but no significant histopathologically detectable disease exacerbations.
Collapse
Affiliation(s)
- Agnes Carolin
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - David Frazer
- Molecular Nutrition, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kexin Yan
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Cameron R Bishop
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Bing Tang
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Wilson Nguyen
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Sheridan L Helman
- Molecular Nutrition, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jay Horvat
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | | | - Daniel J Rawle
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Suhrbier
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- GVN Centre of Excellence, Australian Infectious Disease Research Centre, Brisbane, QLD, Australia
| |
Collapse
|
45
|
Li JW, Mao YM, Chen SL, Ye R, Fei YR, Li Y, Tong SY, Yang HW, He YB. The interplay between metal ions and immune cells in glioma: pathways to immune escape. Discov Oncol 2024; 15:348. [PMID: 39134820 PMCID: PMC11319581 DOI: 10.1007/s12672-024-01229-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
This review explores the intricate roles of metal ions-iron, copper, zinc, and selenium-in glioma pathogenesis and immune evasion. Dysregulated metal ion metabolism significantly contributes to glioma progression by inducing oxidative stress, promoting angiogenesis, and modulating immune cell functions. Iron accumulation enhances oxidative DNA damage, copper activates hypoxia-inducible factors to stimulate angiogenesis, zinc influences cell proliferation and apoptosis, and selenium modulates the tumor microenvironment through its antioxidant properties. These metal ions also facilitate immune escape by upregulating immune checkpoints and secreting immunosuppressive cytokines. Targeting metal ion pathways with therapeutic strategies such as chelating agents and metalloproteinase inhibitors, particularly in combination with conventional treatments like chemotherapy and immunotherapy, shows promise in improving treatment efficacy and overcoming resistance. Future research should leverage advanced bioinformatics and integrative methodologies to deepen the understanding of metal ion-immune interactions, ultimately identifying novel biomarkers and therapeutic targets to enhance glioma management and patient outcomes.
Collapse
Affiliation(s)
- Jin-Wei Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi-Ming Mao
- Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu Province, China
| | - Shi-Liang Chen
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Rui Ye
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi-Ran Fei
- The First Clinical Medical College, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang Province, China
| | - Yue Li
- The First Clinical Medical College, Guangxi Medical University, Nanning, Guangxi Province, China
| | - Shi-Yuan Tong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Hong-Wei Yang
- Department of Clinical Laboratory, Suzhou BOE Hospital, Suzhou, Jiangsu Province, China.
| | - Yi-Bo He
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| |
Collapse
|
46
|
Liu Y, Wu A, Yu B, He J, Yu J, Mao X, Zheng P, Luo Y, Luo J, Pu J, Yan H, Chen D. The influence of iron nutrition on the development of intestine and immune cell divergency in neonatal pigs. J Anim Sci Biotechnol 2024; 15:111. [PMID: 39127747 DOI: 10.1186/s40104-024-01068-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/24/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Appropriate iron supplementation is essential for neonatal growth and development. However, there are few reports on the effects of iron overload on neonatal growth and immune homeostasis. Thus, the aim of this study was to investigate the effects of iron nutrition on neonatal growth and intestinal immunity by administering different levels of iron to neonatal pigs. RESULTS We found that iron deficiency and iron overload resulted in slow growth in neonatal pigs. Iron deficiency and iron overload led to down-regulation of jejunum intestinal barrier and antioxidant marker genes, and promoted CD8+ T cell differentiation in jejunum and mesenteric lymph nodes (MLN) of pigs, disrupting intestinal health. Moreover, iron levels altered serum iron and tissue iron status leading to disturbances in redox state, affecting host innate and adaptive immunity. CONCLUSIONS These findings emphasized the effect of iron nutrition on host health and elucidated the importance of iron in regulating redox state and immunity development. This study provided valuable insights into the regulation of redox state and immune function by iron metabolism in early life, thus contributing to the development of targeted interventions and nutritional strategies to optimize iron nutrition in neonates.
Collapse
Affiliation(s)
- Yao Liu
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Aimin Wu
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Bing Yu
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Jun He
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Jie Yu
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Xiangbing Mao
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Ping Zheng
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Yuheng Luo
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Junqiu Luo
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Junning Pu
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Hui Yan
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.
| | - Daiwen Chen
- Key Laboratory of Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.
| |
Collapse
|
47
|
Li B, Cheng K, Wang T, Peng X, Xu P, Liu G, Xue D, Jiao N, Wang C. Research progress on GPX4 targeted compounds. Eur J Med Chem 2024; 274:116548. [PMID: 38838547 DOI: 10.1016/j.ejmech.2024.116548] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Blocking the System Xc-_ GSH_GPX4 pathway to induce ferroptosis in tumor cells is a novel strategy for cancer treatment. GPX4 serves as the core of the System Xc-/GSH/GPX4 pathway and is a predominant target for inducing ferroptosis in tumor cells. This article summarizes compounds identified in current research that directly target the GPX4 protein, including inhibitors, activators, small molecule degraders, chimeric degraders, and the application of combination therapies with other drugs, aiming to promote further research on the target and related diseases.
Collapse
Affiliation(s)
- Bingru Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Keguang Cheng
- School of Chemistry and Pharmaceutical Sciences, State/Ministry of Education of China Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, China
| | - Tzumei Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xing Peng
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ping Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Guoquan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Dong Xue
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education and School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, China
| | - Ning Jiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing, China
| | - Chao Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| |
Collapse
|
48
|
Jiang Y, Cao Y, Li Y, Bi L, Wang L, Chen Q, Lin Y, Jin H, Xu X, Peng R, Chen Z. SNP alleviates mitochondrial homeostasis dysregulation-mediated developmental toxicity in diabetic zebrafish larvae. Biomed Pharmacother 2024; 177:117117. [PMID: 38996709 DOI: 10.1016/j.biopha.2024.117117] [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: 05/08/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
The incidence of diabetes is increasing annually, and the disease is uncurable due to its complex pathogenesis. Therefore, understanding diabetes pathogenesis and developing new treatments are crucial. This study showed that the NO donor SNP (8 µM) significantly alleviated high glucose-induced developmental toxicity in zebrafish larvae. High glucose levels caused hyperglycemia, leading to oxidative stress and mitochondrial damage from excessive ROS accumulation. This promoted mitochondrial-dependent apoptosis and lipid peroxidation (LPO)-induced ferroptosis, along with immune inflammatory reactions that decreased mitochondrial function and altered intracellular grid morphology, causing imbalanced kinetics and autophagy. After SNP treatment, zebrafish larvae showed improved developmental toxicity and glucose utilization, reduced ROS accumulation, and increased antioxidant activity. The NO-sGC-cGMP signaling pathway, inhibited by high glucose, was significantly activated by SNP, improving mitochondrial homeostasis, increasing mitochondrial count, and enhancing mitochondrial function. It's worth noting that apoptosis, ferroptosis and immune inflammation were effectively alleviated. In summary, SNP improved high glucose-induced developmental toxicity by activating the NO-sGC-cGMP signaling pathway to reduce toxic effects such as apoptosis, ferroptosis and inflammation resulting from mitochondrial homeostasis imbalance.
Collapse
Affiliation(s)
- Yingying Jiang
- Department of Emergency, The Third Affiliated Hospital of Shanghai University, Wenzhou No.3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, China
| | - Yu Cao
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yaoqi Li
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Liuliu Bi
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Lv Wang
- Department of Emergency, The Third Affiliated Hospital of Shanghai University, Wenzhou No.3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, China
| | - Qianqian Chen
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yue Lin
- General Practitioner, The Third Affiliated Hospital of Shanghai University, Wenzhou No.3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, China
| | - Huanzhi Jin
- General Practitioner, The Third Affiliated Hospital of Shanghai University, Wenzhou No.3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, China
| | - Xiaoming Xu
- Scientific Research Center, The Third Affiliated Hospital of Shanghai University, Wenzhou No.3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, China
| | - Renyi Peng
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Zheyan Chen
- Department of Plastic Surgery, The Third Affiliated Hospital of Shanghai University, Wenzhou No.3 Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, China.
| |
Collapse
|
49
|
Xu Y, Huang S, Zhou S, Wang X, Wei M, Chen X, Zong R, Lin X, Li S, Liu Z, Chen Q. Iron Chelator Deferiprone Restores Iron Homeostasis and Inhibits Retinal Neovascularization in Experimental Neovascular Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2024; 65:5. [PMID: 39093298 PMCID: PMC11305424 DOI: 10.1167/iovs.65.10.5] [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: 05/10/2024] [Accepted: 07/10/2024] [Indexed: 08/04/2024] Open
Abstract
Purpose Retinal neovascularization is a significant feature of advanced age-related macular degeneration (AMD) and a major cause of blindness in patients with AMD. However, the underlying mechanism of this pathological neovascularization remains unknown. Iron metabolism has been implicated in various biological processes. This study was conducted to investigate the effects of iron metabolism on retinal neovascularization in neovascular AMD (nAMD). Methods C57BL/6J and very low-density lipoprotein receptor (VLDLR) knockout (Vldlr-/-) mice, a murine model of nAMD, were used in this study. Bulk-RNA sequencing was used to identify differentially expressed genes. Western blot analysis was performed to test the expression of proteins. Iron chelator deferiprone (DFP) was administrated to the mice by oral gavage. Fundus fluorescein angiography was used to evaluate retinal vascular leakage. Immunofluorescence staining was used to detect macrophages and iron-related proteins. Results RNA sequencing (RNA-seq) results showed altered transferrin expression in the retina and RPE of Vldlr-/- mice. Disrupted iron homeostasis was observed in the retina and RPE of Vldlr-/- mice. DFP mitigated iron overload and significantly reduced retinal neovascularization and vascular leakage. In addition, DFP suppressed the inflammation in Vldlr-/- retinas. The reduced signals of macrophages were observed at sites of neovascularization in the retina and RPE of Vldlr-/- mice after DFP treatment. Further, the IL-6/JAK2/STAT3 signaling pathway was activated in the retina and RPE of Vldlr-/- mice and reversed by DFP treatment. Conclusions Disrupted iron metabolism may contribute to retinal neovascularization in nAMD. Restoring iron homeostasis by DFP could be a potential therapeutic approach for nAMD.
Collapse
Affiliation(s)
- Yuan Xu
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shiya Huang
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shengmei Zhou
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xin Wang
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Mingyan Wei
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaodong Chen
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Rongrong Zong
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiang Lin
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Shiying Li
- Department of Ophthalmology, the First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Zuguo Liu
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Qian Chen
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
50
|
Hajar A, Swathi NL, Ali A. Immunological Insights Into Nutritional Deficiency Disorders. ADVANCES IN MEDICAL DIAGNOSIS, TREATMENT, AND CARE 2024:61-84. [DOI: 10.4018/979-8-3693-2947-4.ch004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Essential nutrients play a vital role in influencing immune cell development. This chapter explores the crucial relationship between nutrition and the immune system, delving into the profound impact of dietary choices on overall health. Research highlights the benefits of nutrient-rich foods in supporting optimal immune function, while deficiencies in key nutrients (vitamins A, D, zinc, and iron) compromise immune responses, increasing susceptibility to infections. The bidirectional nature of the relationship is emphasized, underscoring the critical role of a balanced diet in supporting immune cell development, activation, and function. Case studies illustrate immunological vulnerabilities linked to inadequate nutritional status, stressing the importance of maintaining optimal nutrient levels for a robust immune system. In summary, an individual's nutritional status significantly influences immune response effectiveness. Addressing deficiencies through supplementation, dietary interventions, and public health initiatives is crucial for improving immune function.
Collapse
Affiliation(s)
- Azraida Hajar
- Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco
| | - N. L. Swathi
- Sri Venkateswara College of Pharmacy, Jawaharlal Nehru Technological University, India
| | - Awais Ali
- Abdul Wali Khan University, Mardan, Pakistan
| |
Collapse
|