1
|
Gao Q, Zhou Y, Chen Y, Hu W, Jin W, Zhou C, Yuan H, Li J, Lin Z, Lin W. Role of iron in brain development, aging, and neurodegenerative diseases. Ann Med 2025; 57:2472871. [PMID: 40038870 PMCID: PMC11884104 DOI: 10.1080/07853890.2025.2472871] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
It is now understood that iron crosses the blood-brain barrier via a complex metabolic regulatory network and participates in diverse critical biological processes within the central nervous system, including oxygen transport, energy metabolism, and the synthesis and catabolism of myelin and neurotransmitters. During brain development, iron is distributed throughout the brain, playing a pivotal role in key processes such as neuronal development, myelination, and neurotransmitter synthesis. In physiological aging, iron can selectively accumulate in specific brain regions, impacting cognitive function and leading to intracellular redox imbalance, mitochondrial dysfunction, and lipid peroxidation, thereby accelerating aging and associated pathologies. Furthermore, brain iron accumulation may be a primary contributor to neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. Comprehending the role of iron in brain development, aging, and neurodegenerative diseases, utilizing iron-sensitive Magnetic Resonance Imaging (MRI) technology for timely detection or prediction of abnormal neurological states, and implementing appropriate interventions may be instrumental in preserving normal central nervous system function.
Collapse
Affiliation(s)
- Qiqi Gao
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiyang Zhou
- Department of Urology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yu Chen
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Hu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenwen Jin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunting Zhou
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hao Yuan
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianshun Li
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenlang Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
2
|
Qiu Q, Sun Q, Yang J, Yuan Q, Wang P, Liu Q, Cui Z, Ma X, Li M. The molecular mechanism by which CTSB degrades FPN to disrupt macrophage iron homeostasis and promote the progression of atherosclerosis. Mol Cell Biochem 2025; 480:3889-3906. [PMID: 39960586 DOI: 10.1007/s11010-025-05228-9] [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: 11/25/2024] [Accepted: 02/05/2025] [Indexed: 05/23/2025]
Abstract
The incidence of atherosclerosis (AS) remains high, and iron-dependent cell death (termed ferroptosis) is thought to play a key role in the progression of AS. Studies have shown that cathepsin B (CTSB) is an important regulatory molecule in atherosclerosis. However, how CTSB regulates AS progression has not been reported, and whether it is related to ferroptosis is poorly studied. In the present study, we observed a significant upregulation of CTSB expression in two AS models, ApoE knockout mice and SD rats given a HFD. According to our findings, CTSB can promote development of the AS plaque region, while inhibition of CTSB showed a reduction of AS lesion area and lipid deposition. Single-cell transcriptome analysis of AS tissue from humans revealed that CTSB is primarily expressed in macrophages. Oxidized low-density lipoprotein (ox-LDL) significantly enhanced macrophage CTSB expression, and induced ferroptosis in vitro. Mechanistically, Ferroportin (FPN) is the binding target of CTSB. CTSB can negatively regulate the protein level of FPN and promote its degradation, which further leads to ferroptosis of macrophages. We confirmed that ferroptosis in macrophages could be effectively inhibited by knockdown or pharmacological inhibition of CTSB, which in turn slowed the progression of AS. In conclusion, our study suggests that CTSB disrupts iron homeostasis in macrophages by degrading FPN and induces ferroptosis, thereby exacerbating the development of AS. Targeting CTSB may become an important potential strategy for the treatment of AS.
Collapse
Affiliation(s)
- Quanli Qiu
- Jinzhou Medical University Graduate Training Base (PLA 960, Hospital), 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Qiyu Sun
- Jinzhou Medical University Graduate Training Base (PLA 960, Hospital), 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Jiaxin Yang
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Qingxin Yuan
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Ping Wang
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Qingwei Liu
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Zhenzhen Cui
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Xiaowen Ma
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
| | - Min Li
- Jinzhou Medical University Graduate Training Base (PLA 960, Hospital), 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
| |
Collapse
|
3
|
Zhu D, Wu JZ, Griffin PT, Samuelson BA, Sinclair DA, Kane AE. Metabolomics biomarkers of frailty: a longitudinal study of aging female and male mice. NPJ AGING 2025; 11:40. [PMID: 40410187 PMCID: PMC12102153 DOI: 10.1038/s41514-025-00237-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025]
Abstract
Frailty is an age-related geriatric syndrome. We performed a longitudinal study of aging female (n = 40) and male (n = 47) C57BL/6NIA mice, measured frailty index and derived metabolomics data from plasma. We identify age-related differentially abundant metabolites, determine frailty-related metabolites, and generate frailty features, both in the whole cohort and sex-stratified subgroups. Using the features, we perform an association study and build a metabolomics-based frailty clock. We find that frailty-related metabolites are enriched for amino acid metabolism and metabolism of cofactors and vitamins, include ergothioneine, tryptophan and alpha-ketoglutarate, and present sex dimorphism. We identify B vitamin metabolism related flavin-adenine dinucleotide and pyridoxate as female-specific frailty biomarkers, and lipid metabolism related sphingomyelins, glycerophosphoethanolamine and glycerophosphocholine as male-specific frailty biomarkers. These associations are confirmed in a validation cohort, with ergothioneine and perfluorooctanesulfonate identified as robust frailty biomarkers. Our results identify sex-specific metabolite frailty biomarkers, and shed light on potential mechanisms.
Collapse
Affiliation(s)
- Dantong Zhu
- Institute for Systems Biology, Seattle, WA, USA
| | - Judy Z Wu
- Institute for Systems Biology, Seattle, WA, USA
| | - Patrick T Griffin
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA, USA
| | | | - David A Sinclair
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA, USA
| | - Alice E Kane
- Institute for Systems Biology, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
4
|
Di Lorenzo R, Marzetti E, Coelho-Junior HJ, Calvani R, Pesce V, Landi F, Leeuwenburgh C, Picca A. Iron Metabolism and Muscle Aging: Where Ferritinophagy Meets Mitochondrial Quality Control. Cells 2025; 14:672. [PMID: 40358196 PMCID: PMC12072144 DOI: 10.3390/cells14090672] [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: 04/04/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
In older adults with reduced physical performance, an increase in the labile iron pool within skeletal muscle is observed. This accumulation is associated with an altered expression of mitochondrial quality control (MQC) markers and increased mitochondrial DNA damage, supporting the hypothesis that impaired MQC contributes to muscle dysfunction during aging. The autophagy-lysosome system plays a critical role in MQC by tagging and engulfing proteins and organelles for degradation in lysosomes. The endolysosomal system is also instrumental in transferrin recycling, which, in turn, regulates cellular iron uptake. In the neuromuscular system, the autophagy-lysosome system supports the structural integrity of neuromuscular junctions, and its dysfunction contributes to muscle atrophy. While MQC was thought to protect against iron-induced cell death, the discovery of ferroptosis, a form of iron-dependent cell death, has highlighted a complex interplay between MQC and iron-inflicted damage. Ferritinophagy, the autophagic degradation of ferritin, if overactivated, can induce ferroptosis. Alternatively, aging may impair ferritinophagy, leading to ferritin accumulation and the release of toxic labile iron under stress, exacerbating oxidative damage and cellular senescence. Physical activity supports muscle health also by preserving mitochondrial quantity and quality and enhancing bioenergetics. However, therapeutic strategies for preventing or reversing physical function decline in aging are still lacking due to the insufficient understanding of the underlying mechanisms. Unveiling how disruptions in iron homeostasis impact muscle quality in older adults may allow for the development of therapeutic strategies targeting iron handling to alleviate age-associated muscle decline.
Collapse
Affiliation(s)
- Rosa Di Lorenzo
- Department of Biosciences, Biotechnologies, and Environment, Università degli Studi di Bari Aldo Moro, Via Edoardo Orabona 4, 70125 Bari, Italy; (R.D.L.); (V.P.)
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (R.C.); (F.L.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
| | - Helio José Coelho-Junior
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
| | - Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (R.C.); (F.L.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
| | - Vito Pesce
- Department of Biosciences, Biotechnologies, and Environment, Università degli Studi di Bari Aldo Moro, Via Edoardo Orabona 4, 70125 Bari, Italy; (R.D.L.); (V.P.)
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (R.C.); (F.L.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
| | - Christiaan Leeuwenburgh
- Department of Physiology and Aging, University of Florida, 2004 Mowry Road, Gainesville, FL 32611, USA
| | - Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
- Department of Medicine and Surgery, LUM University, Str. Statale 100, 70010 Casamassima, Italy
| |
Collapse
|
5
|
Cao S, Pang Y, Wei Y, Wang D, Xiong A, Yan J, Zeng H. Bibliometric and graphical analysis of ferroptosis and aging research: Trends, gaps, and future directions. Pathol Res Pract 2025; 269:155949. [PMID: 40174280 DOI: 10.1016/j.prp.2025.155949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Over the past 12 years, a significant body of evidence derived from extensive research has underscored the pivotal involvement of ferroptosis in the mechanisms underlying aging. Despite the growing body of literature on this topic, there remains a paucity of analytical and descriptive studies that explore its trajectory, key research directions, current trends, primary focal points, and future outlooks. This research endeavors to provide an exhaustive overview of the advancements in understanding the relationship between ferroptosis and aging over the past 12 years. The dataset utilized in this study was extracted from the Web of Science, encompassing records from January 1, 2012, through June 19, 2024. We conducted comprehensive bibliometric and visual analyses using advanced analytical tools. The results highlight China's dominant contribution, which accounts for 48.52 % of total publications, positioning it as a key player in this research area. Leading institutions, including Columbia University, Southern Medical University, and the Salk Institute for Biological Studies, demonstrate high research productivity. Pamela Maher and Gu Wei are identified as the most prolific researchers in this field. Free Radical Biology and Medicine is the leading journal, publishing the most articles in this field. This study identifies mitochondrial diseases, arrhythmias, Parkinson's disease, hepatocellular carcinoma, and iron-refractory iron deficiency anemia as the key diseases investigated in this field. This bibliometric evaluation offers critical perspectives for both experienced scholars and early-career researchers, enabling the identification of novel ideas and advancements within this domain.
Collapse
Affiliation(s)
- Siyang Cao
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China
| | - Yingchen Pang
- Department of Pulmonary and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Pulmonary and Critical Care Medicine, Shenzhen Xinhua Hospital, Shenzhen, Guangdong, PR China
| | - Yihao Wei
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong; Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, PR China; Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong, PR China
| | - Deli Wang
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China
| | - Ao Xiong
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China.
| | - Jun Yan
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China.
| | - Hui Zeng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, PR China.
| |
Collapse
|
6
|
Liu YJ, Jia GR, Zhang SH, Guo YL, Ma XZ, Xu HM, Xie JX. The role of microglia in neurodegenerative diseases: from the perspective of ferroptosis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01560-4. [PMID: 40307457 DOI: 10.1038/s41401-025-01560-4] [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: 12/17/2024] [Accepted: 04/06/2025] [Indexed: 05/02/2025]
Abstract
Iron plays a pivotal role in numerous fundamental biological processes in the brain. Among the various cell types in the central nervous system, microglia are recognized as the most proficient cells in accumulating and storing iron. Nonetheless, iron overload can induce inflammatory phenotype of microglia, leading to the production of proinflammatory cytokines and contributing to neurodegeneration. A growing body of evidence shows that disturbances in iron homeostasis in microglia is associated with a range of neurodegenerative disorders. Recent research has revealed that microglia are highly sensitive to ferroptosis, a form of iron-dependent cell death. How iron overload influences microglial function? Whether disbiosis in iron metabolism and ferroptosis in microglia are involved in neurodegenerative disorders and the underlying mechanisms remain to be elucidated. In this review we focus on the recent advances in research on microglial iron metabolism as well as ferroptosis in microglia. Meanwhile, we provide a comprehensive overview of the involvement of microglial ferroptosis in neurodegenerative disorders from the perspective of crosstalk between microglia and neuron, with a focus on Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Ying-Juan Liu
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
| | - Guo-Rui Jia
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Sheng-Han Zhang
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Yun-Liang Guo
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
| | - Xi-Zhen Ma
- College of Life Sciences and Health, University of Health and Rehabilitation Science, Qingdao, 266113, China.
| | - Hua-Min Xu
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Jun-Xia Xie
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
7
|
Khanal V, Carroll M, Carter J, Zhong Y, Chikkamagaluru S, Sato A, Allen R, Wankhade U, Dole N. Lipocalin-2 Regulates Osteocyte Ferroptosis and Osteocyte-Osteoblast Crosstalk via Wnt Signaling to Control Bone Formation. RESEARCH SQUARE 2025:rs.3.rs-6430607. [PMID: 40343339 PMCID: PMC12060985 DOI: 10.21203/rs.3.rs-6430607/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Osteoporosis is a multifactorial disease, and emerging evidence suggests that iron overload contributes to its progression. Here, we identify Lipocalin-2 (LCN2), a cytokine secreted by bone cells with endocrine effects on other tissues, as a local regulator of osteocyte iron metabolism and a mediator of skeletal deterioration. Our findings reveal that LCN2 promotes iron accumulation, mitochondrial dysfunction, and ferroptosis in osteocytes in a process dependent on LCN2 receptor SLC22A17. Genetic ablation of Lcn2 (Dmp1-Cre; Lcn2 fl/fl ) in osteocytes mitigates their ferroptotic vulnerability by preserving mitochondrial integrity and limiting iron overload. Remarkably, LCN2 deletion enhances osteocyte dendricity and lacunocanalicular network, supporting their function in bone remodeling. Mechanistically, we demonstrate that Lcn2 ablation in osteocytes decreases DKK1 and SOST expression in bone, leading to increased Wnt/β-catenin signaling and osteoblast-driven bone formation. Using in vitro and in vivo approaches, we establish the LCN2-SLC22A17 axis as a key pathway linking iron homeostasis, osteocyte dysfunction, and skeletal remodeling. These findings provide insight into a previously unrecognized mechanism underlying iron-driven bone loss and suggest that targeting LCN2 could offer therapeutic potential for osteoporosis.
Collapse
Affiliation(s)
| | | | | | - Ying Zhong
- University of Arkansas for Medical Sciences
| | | | - Amy Sato
- University of Arkansas for Medical Sciences
| | - Ryan Allen
- University of Arkansas for Medical Sciences
| | | | - Neha Dole
- University of Arkansas for Medical Sciences
| |
Collapse
|
8
|
Saeed BI, Uthirapathy S, Kubaev A, Ganesan S, Shankhyan A, Gupta S, Joshi KK, Kariem M, Jasim AS, Ahmed JK. Ferroptosis as a key player in the pathogenesis and intervention therapy in liver injury: focusing on drug-induced hepatotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04115-w. [PMID: 40244448 DOI: 10.1007/s00210-025-04115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Globally, drug-induced hepatotoxicity or drug-induced liver injury (DILI) is a serious clinical concern. Knowing the processes and patterns of cell death is essential for finding new therapeutic targets since there are not many alternatives to therapy for severe liver lesions. Excessive lipid peroxidation is a hallmark of ferroptosis, an iron-reliant non-apoptotic cell death linked to various liver pathologies. When iron is pathogenic, concomitant inflammation may exacerbate iron-mediated liver injury, and the hepatocyte necrosis that results is a key element in the fibrogenic response. The idea that dysregulated metabolic pathways and compromised iron homeostasis contribute to the development of liver injury by ferroptosis is being supported by new data. Various ferroptosis-linked genes and pathways have been linked to liver injury, although the molecular processes behind ferroptosis's pathogenicity are not well known. Here, we delve into the features of ferroptosis, the processes governing ferroptosis, and our current knowledge of iron metabolism. We also provide an overview of ferroptosis's involvement in the pathophysiology of liver injury, particularly DILI. Lastly, the therapeutic possibilities of ferroptosis targeting for liver injury management have been provided. Natural products, nanoparticles (NPs), mesenchymal stem cell (MSC), and their exosomes have attracted increasing attention among such therapeutics.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
9
|
Zhang Q, Zhang Y, Zhang S, Zeng Y, He Z, Yang K, Luo J, Guo H, Chi B. Elucidation of the interaction between apo-transferrin and indisulam via multi-spectroscopic techniques and molecular modeling. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 330:125652. [PMID: 39736258 DOI: 10.1016/j.saa.2024.125652] [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: 08/20/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/01/2025]
Abstract
Apo-transferrin (apo-TRF) is a vital protein for maintaining iron balance in the body, which is produced by the liver. Indisulam (IDM) has been extensively used to treat cancer in clinical study and has been identified as a molecular glue. Iron imbalances in the body are believed to encourage the growth and spread of cancer cells. Thus, understanding the interactions between apo-TRF and IDM may serve as a foundation for identifying novel therapeutic strategies for cancer associated with iron imbalances. In this study, multi-spectroscopic methods and computer simulations were employed to explore the binding mode between apo-TRF and IDM, as well as to investigate IDM's impact on the biological functions of apo-TRF. Multi-spectroscopic studies indicated that IDM and apo-TRF formed binary complexes with Ka of 1.274 × 104 M-1 at 298 K. The H-bonds and van der Waals forces were the dominant interaction forces based on an analysis of the thermodynamic parameters (ΔHθ = -37.565 kJ/mol, ΔSθ = -46.665 J mol-1 K-1). Three-dimensional (3D) and circular dichroism (CD) spectra revealed the conformational of apo-TRF changed by IDM, resulting in a looser and more unfolded structure. With escalating concentrations of IDM, a notable reduction in the binding affinity between apo-TRF and Fe3+ was observed, indicating that IDM could potentially alter iron transfer mediated by apo-TRF. Molecular docking analysis indicated that IDM docked in the apo-TRF iron-binding pocket. After in-depth analysis of the molecular dynamic results, it was found that Asp392 played an important role in this interaction. In addition, accessible surface area (ASA) values of key residues (Tyrosine, Aspartate, and Histidine) for iron transfer were altered, which could be a possible reason for the change in iron transport.
Collapse
Affiliation(s)
- Qiumei Zhang
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Yue Zhang
- School of Pharmacy, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Shuyuan Zhang
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Yujing Zeng
- School of Pharmacy, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Zimeng He
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Kaiyu Yang
- School of Pharmacy, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Jiaqing Luo
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Hui Guo
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Baozhu Chi
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China.
| |
Collapse
|
10
|
Bolesławska I, Bolesławska-Król N, Jakubowski K, Przysławski J, Drzymała-Czyż S. Lactoferrin-A Regulator of Iron Homeostasis and Its Implications in Cancer. Molecules 2025; 30:1507. [PMID: 40286136 PMCID: PMC11990823 DOI: 10.3390/molecules30071507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
Cancer is a global health challenge, and its development is closely linked to iron metabolism. Cancer cells have an increased demand for this element, which promotes their proliferation, invasion, and metastasis. Excess iron catalyzes the formation of reactive oxygen species (ROS), which can both induce ferroptosis and initiate oncogenic signaling pathways. The deregulation of iron metabolism in cancer patients leads to anemia or toxic iron overload and also affects the gut microbiota. Lactoferrin (LF), a glycoprotein with strong iron chelating properties, can regulate its availability to cancer cells, thereby limiting their growth and progression. By chelating free Fe ions, LF reduces oxidative stress and inhibits the mechanisms that promote carcinogenesis. Additionally, it exhibits immunomodulatory and anti-inflammatory effects and may enhance the body's anti-tumor response. This review analyses the mechanisms of action of lactoferrin in the context of cancer, with a particular focus on its chelating, antioxidant, and immunomodulatory properties. The multidirectional effects of LF make it a promising component of preventive and therapeutic strategies, requiring further clinical studies.
Collapse
Affiliation(s)
- Izabela Bolesławska
- Department of Bromatology, Poznan University of Medical Sciences, 60-806 Poznan, Poland; (K.J.); (J.P.); (S.D.-C.)
| | - Natasza Bolesławska-Król
- Student Society of Radiotherapy, Collegium Medicum, University of Zielona Góra, Zyta 28, 65-046 Zielona Góra, Poland;
| | - Karol Jakubowski
- Department of Bromatology, Poznan University of Medical Sciences, 60-806 Poznan, Poland; (K.J.); (J.P.); (S.D.-C.)
| | - Juliusz Przysławski
- Department of Bromatology, Poznan University of Medical Sciences, 60-806 Poznan, Poland; (K.J.); (J.P.); (S.D.-C.)
| | - Sławomira Drzymała-Czyż
- Department of Bromatology, Poznan University of Medical Sciences, 60-806 Poznan, Poland; (K.J.); (J.P.); (S.D.-C.)
| |
Collapse
|
11
|
Yi T, Wu S, Yang Y, Li X, Yang S, Zhang Y, Zhang L, Hu Y, Zhang G, Li J, Yang D. Single-nucleus RNA sequencing reveals dynamic changes in the microenvironment of visceral adipose tissue and metabolic characteristics after cold exposure. Front Endocrinol (Lausanne) 2025; 16:1562431. [PMID: 40196457 PMCID: PMC11973077 DOI: 10.3389/fendo.2025.1562431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Visceral adipose tissue (VAT) plays a crucial role in regulating systemic metabolic balance. Excess accumulation of VAT is closely associated with various metabolic disorders, a process that involves the coordinated actions of multiple cell types within the tissue. Cold exposure, as a potential intervention, has been proposed to improve metabolic dysfunction. However, the heterogeneity of VAT and its comprehensive metabolic characteristics under cold exposure remain unclear. Methods We collected epididymal white adipose tissue (eWAT) of C57BL/6J mice after cold exposure at three different time points for single-nucleus RNA sequencing (snRNA-seq) analysis. Results We successfully identified ten major cell types in eWAT, enabling understanding of the dynamic changes in the eWAT microenvironment and its metabolic features during cold exposure. This study revealed that cold exposure for 1 day reduced cellular metabolic activity and intercellular communication in eWAT including receptor-ligand-based cell communication and metabolite-mediated interactions. However, after 14 days of cold acclimation, the metabolic activity of adipocytes was significantly enhanced, and intercellular metabolic communication was restored. Additionally, prolonged cold exposure promoted the formation of a distinct adipocyte subpopulation that may be associated with UCP1-independent thermogenesis. These changes may be a new homeostatic state established by VAT to adapt to the cold environment. The study also identified the importance of adipocytes, adipose stem and progenitor cells, myeloid cells, and endothelial cells in the process of cold adaptation. Discussion This research provides valuable insights into the cellular heterogeneity, adipocyte remodeling, and metabolic reprogramming in eWAT after cold exposure. It highlights the critical role of transcriptional dynamics in eWAT during cold exposure and provides new perspectives on the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Ting Yi
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Shuai Wu
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Yusha Yang
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Xi Li
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Shuran Yang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yongqiang Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Li Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Yuyu Hu
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Guanyu Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Jun Li
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Danfeng Yang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| |
Collapse
|
12
|
Zhang T, Zhang Y, Xie J, Lu D, Wang L, Zhao S, Zhou J, Cheng Y, Kou T, Wang J, Chen Y, Xu L, Hu X, Ying Y, Wang J, Xin X, Xu X, Lei S, Qiu C, Wu J, Lyu Q, Cao T. Ferroptosis in neurodegenerative diseases: mechanisms and therapeutic potential of stem cell derivatives. Front Cell Dev Biol 2025; 13:1577382. [PMID: 40191227 PMCID: PMC11968680 DOI: 10.3389/fcell.2025.1577382] [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: 02/15/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Ferroptosis, a non-apoptotic, iron-dependent form of regulated cell death, is closely related to the pathogenesis of neurodegenerative diseases. Stem cells and their derivatives exhibit remarkable potential in modulating ferroptosis, offering promising therapeutic intervention for neurodegenerative diseases. In this review, we systematically explore neurological aging and its association with cognitive impairment and neurodegenerative diseases, with focus on the molecular mechanisms of ferroptosis in neurodegenerative diseases and the potential therapeutic strategies of stem cell derivatives for neurological diseases.
Collapse
Affiliation(s)
- Ting Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yusu Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinpeng Xie
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dandan Lu
- School and hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lihong Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuaifei Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Zhou
- School and hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Stomatology the Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yang Cheng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ting Kou
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jue Wang
- School and hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangyu Hu
- School and hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxiu Ying
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Wang
- School and hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoshuang Xin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xu Xu
- School and hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Siyun Lei
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| | - Chenyu Qiu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| | - Jinhua Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiqi Lyu
- Department of Neuroradiology, Singapore General Hospital, Singapore, Singapore
| | - Tong Cao
- School and hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| |
Collapse
|
13
|
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
|
14
|
Li Y, Li Y, Li P, Yang L, Li H. 4-Octyl Itaconate Attenuates Postmenopausal Osteoporosis by Inhibiting Ferroptosis and Enhancing Osteogenesis via the Nrf2 Pathway. Inflammation 2025:10.1007/s10753-025-02268-7. [PMID: 39984770 DOI: 10.1007/s10753-025-02268-7] [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: 11/12/2024] [Revised: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) play an important role in bone metabolism and tissue repair, and their ability to differentiate into osteoblasts is crucial in the treatment of bone diseases such as postmenopausal osteoporosis (PMOP). However, the function of BMSCs may be affected by ferroptosis. Ferroptosis is a cell death mode characterized by excess Fe2+ and lipid peroxidation, which significantly affects the survival rate and differentiation ability of BMSCs. This study investigated the effect of exogenous itaconate derivative 4-octyl itaconate (4-OI) on Erastin-induced BMSCs ferroptosis. The results showed that 4-OI significantly inhibited Erastin-induced BMSCs ferroptosis by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway, reduced reactive oxygen species levels and oxidative damage, and restored antioxidant capacity. At the same time, 4-OI promoted the osteogenic differentiation of BMSCs. Further experiments showed that Nrf2-IN-1, an inhibitor of the Nrf2 pathway, could reverse the protective effect of 4-OI. In vivo, 4-OI was shown to reduce bone loss in ovariectomized (OVX) mice, as assessed by Micro-CT analysis. Immunofluorescence staining further revealed increased GPX4 and Nrf2 expression in vertebral tissues following 4-OI treatment. These results indicate that 4-OI improves ferroptosis of BMSCs and enhances osteogenic differentiation ability by activating the Nrf2 pathway, providing new research ideas and potential targets for the treatment of PMOP.
Collapse
Affiliation(s)
- You Li
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Yang Li
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Pengfei Li
- School of Postgraduate, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Yang
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China.
| | - Haijun Li
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China.
- School of Postgraduate, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
15
|
Cilleros-Holgado P, Gómez-Fernández D, Piñero-Pérez R, Romero-Domínguez JM, Reche-López D, Álvarez-Córdoba M, Romero-González A, López-Cabrera A, De Oliveira MC, Rodríguez-Sacristán A, González-Granero S, García-Verdugo JM, Sánchez-Alcázar JA. Polydatin and Nicotinamide Prevent Iron Accumulation and Lipid Peroxidation in Cellular Models of Mitochondrial Diseases. Antioxidants (Basel) 2025; 14:215. [PMID: 40002401 PMCID: PMC11851670 DOI: 10.3390/antiox14020215] [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: 12/22/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Ferroptosis, an iron-dependent form of non-apoptotic cell death, is regulated by a complex network involving lipid metabolism, iron homeostasis, and the oxidative-reductive system, with iron accumulation and lipid peroxidation as key drivers. Mitochondrial dysfunction and ROS overproduction often underlie the pathogenesis of mitochondrial diseases, for which treatment options are limited, emphasizing the need for novel therapies. In this study, we investigated whether polydatin and nicotinamide could reverse ferroptosis-related pathological features in cellular models derived from patients with pathogenic GFM1 variants. Mutant fibroblasts showed increased iron and lipofuscin accumulation, altered expression of iron metabolism-related proteins, elevated lipid peroxidation, and heightened susceptibility to erastin-induced ferroptosis. Treatment with polydatin and nicotinamide effectively corrected these alterations and reduced iron accumulation and lipid peroxidation in induced neurons. Furthermore, chloramphenicol treatment in control cells mimicked the mutant phenotype, suggesting that these pathological changes are linked to the mitochondrial protein synthesis defect characteristic of pathogenic GFM1 variants. Notably, adding vitamin E to the polydatin and nicotinamide co-treatment resulted in a reduction in the minimum effective concentration, suggesting potential benefits of its inclusion. In conclusion, the combination of polydatin, nicotinamide, and vitamin E could represent a promising therapeutic option for patients with mitochondrial disorders caused by pathogenic GFM1 variants.
Collapse
Affiliation(s)
- Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - David Gómez-Fernández
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - José Manuel Romero-Domínguez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Ana Romero-González
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Alejandra López-Cabrera
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Marta Castro De Oliveira
- Neuropediatria, Neurolinkia, C. Jardín de la Isla, 8, Local 4 y 5, 41014 Sevilla, Spain;
- FEA Pediatría, Centro Universitario Hospitalar de Faro, R. Leão Penedo, 8000-386 Faro, Portugal
| | - Andrés Rodríguez-Sacristán
- Neuropediatría, Servicio de Pediatría, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain;
- Departamento de Farmacología, Radiología y Pediatría de la Facultad de Medicina de la Universidad de Sevilla, 41009 Sevilla, Spain
| | - Susana González-Granero
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46980 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46980 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José Antonio Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| |
Collapse
|
16
|
Song Y, Hai E, Zhang N, Zhang Y, Wang J, Han X, Zhang J. Oocyte transcriptomes and follicular fluid proteomics of ovine atretic follicles reveal the underlying mechanisms of oocyte degeneration. BMC Genomics 2025; 26:97. [PMID: 39893388 PMCID: PMC11786490 DOI: 10.1186/s12864-025-11291-9] [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: 05/27/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND In mammals, female fertility is influenced by the result of follicular development (ovulation or atresia). Follicular atresia is a complex physiological process that results in the degeneration of oocytes from the ovary. However, the molecular mechanisms of oocyte degeneration and key protein markers of follicular atresia remain unclear. In this study, we investigated the complex transcriptional regulatory mechanisms and protein profiles in oocytes and follicular fluid in atretic follicle stages using single-cell RNA sequencing and tandem mass tag proteomics. RESULTS First, through paired analysis of different follicle development stages, we identified 175 atresia-specific genes and eight candidate oocyte-secreted factors, including PKG1, YTHDF2, and MYC. Meanwhile, we also characterized unique features of the oocyte transcriptional landscape in the atretic follicle stage that displayed cell death-related transcriptional changes and mechanisms, such as autophagy (TBK1 and IRS4), necroptosis (PKR), and apoptosis (MARCKS). Moreover, we identified atresia-specific genes, namely FTH1, TF, and ACSL4, which may participate in regulation of oocyte ferroptosis in atretic follicles through a series of mechanisms including ferritinophagy, ferritin transport, and lipid metabolism. Additionally, we uncovered 333 differentially expressed proteins that may coordinate follicular atresia and revealed key pathways, such as negative regulation of angiogenesis, metabolic pathways, and transcription and mRNA splicing, that lead to oocyte degeneration. Finally, by combining transcriptome and proteomics analyses, we identified two oocyte-secreted biomarkers, PGK1 and ANGPT2, that may be associated with follicular atresia. CONCLUSIONS In conclusion, our work offers a thorough characterization of oocyte transcription mechanism and follicular fluid protein changes in ovine atretic follicles, which offers a crucial reference for analyzing the mechanism of follicular atresia and establishing an oocyte quality assessment system in sheep.
Collapse
Affiliation(s)
- Yukun Song
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Erhan Hai
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Nan Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Yu Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Junlan Wang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Xitong Han
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Jiaxin Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China.
| |
Collapse
|
17
|
Huang B, Wen W, Ye S. Iron-Deficiency Anemia Elevates Risk of Diabetic Kidney Disease in Type 2 Diabetes Mellitus. J Diabetes 2025; 17:e70060. [PMID: 39968673 PMCID: PMC11836615 DOI: 10.1111/1753-0407.70060] [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: 08/22/2024] [Revised: 12/02/2024] [Accepted: 02/01/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE This study aims to explore the link between iron deficiency anemia (IDA) and diabetic kidney disease (DKD) and assess the safety of iron supplementation. It also investigates key mechanisms and molecules involved in iron deficiency's role in disease development. METHODS A retrospective analysis was conducted on 1,398 T2DM patients using propensity score matching to identify risk factors for DKD. Mendelian randomization (MR) was used to explore causal relationships between IDA, iron supplementation, liver iron content, and DKD. The GSE27999 dataset was analyzed to examine how an iron-deficient diet affects kidney-related gene expression. Key pathways and molecules were identified through GSEA, GO/KEGG, and PPI analysis. RESULTS Retrospective data showed a correlation between hemoglobin levels and DKD risk. Logistic regression confirmed that IDA increased DKD risk independently of other factors. MR revealed a causal link between IDA and DKD, with no significant effect from iron supplementation. GSE27999 analysis identified 580 differentially expressed genes, enriched in pathways like cytokine signaling, oxidative biology, and small molecule transport. PPI analysis highlighted 10 key hub genes, including Cyp2d26 and Fgf4. CONCLUSION IDA increases susceptibility to DKD, possibly through oxidative stress and altered small molecule transport. However, iron supplementation does not appear to increase the risk of DKD.
Collapse
Affiliation(s)
- Bin Huang
- Department of EndocrinologyThe First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of ChinaHefeiAnhuiChina
| | - Wenjie Wen
- Anhui Province Engineering Research Center for Dental Materials and Application, School of StomatologyWannan Medical CollegeWuhuChina
| | - Shandong Ye
- Department of EndocrinologyThe First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of ChinaHefeiAnhuiChina
| |
Collapse
|
18
|
Zhu D, Wu JZ, Griffin P, Samuelson BA, Sinclair DA, Kane AE. Metabolomics biomarkers of frailty: a longitudinal study of aging female and male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634160. [PMID: 39896479 PMCID: PMC11785134 DOI: 10.1101/2025.01.22.634160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Frailty is an age-related geriatric syndrome, for which the mechanisms remain largely unknown. We performed a longitudinal study of aging female (n = 40) and male (n = 47) C57BL/6NIA mice, measured frailty index and derived metabolomics data from plasma samples. We identify differentially abundant metabolites related to aging, determine frailty related metabolites via a machine learning approach, and generate a union set of frailty features, both in the whole cohort and in sex-stratified subgroups. Using the features, we perform an association study and build a metabolomics-based frailty clock. We find that frailty related metabolites are enriched for amino acid metabolism and metabolism of cofactors and vitamins, include ergothioneine, tryptophan, and alpha-ketoglutarate, and present sex dimorphism. We identify B vitamin metabolism related flavin adenine dinucleotide and pyridoxate as female-specific frailty biomarkers, and lipid metabolism related sphingomyelins, glycerophosphoethanolamine and glycerophosphocholine as male-specific frailty biomarkers. These associations are confirmed in a validation cohort, with ergothioneine and perfluorooctanesulfonate identified as robust frailty biomarkers. In summary, our results identify sex-specific metabolite biomarkers of frailty in aging, and shed light on potential mechanisms involved in frailty.
Collapse
Affiliation(s)
- Dantong Zhu
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Judy Z. Wu
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Patrick Griffin
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA 02115, USA
| | | | - David A. Sinclair
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA 02115, USA
| | - Alice E. Kane
- Institute for Systems Biology, Seattle, WA 98109, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
19
|
Ju JJ, Hang LH. Neuroinflammation and iron metabolism after intracerebral hemorrhage: a glial cell perspective. Front Neurol 2025; 15:1510039. [PMID: 39882361 PMCID: PMC11774705 DOI: 10.3389/fneur.2024.1510039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is the most common subtype of hemorrhagic stroke causing significant morbidity and mortality. Previously clinical treatments for ICH have largely been based on a single pathophysiological perspective, and there remains a lack of curative interventions. Following the rupture of cerebral blood vessels, blood metabolites activate resident immune cells such as microglia and astrocytes, and infiltrate peripheral immune cells, leading to the release of a series of inflammatory mediators. Degradation of hemoglobin produces large amounts of iron ions, leading to an imbalance of iron homeostasis and the production of large quantities of harmful hydroxyl radicals. Neuroinflammation and dysregulation of brain iron metabolism are both important pathophysiological changes in ICH, and both can exacerbate secondary brain injury. There is an inseparable relationship between brain iron metabolism disorder and activated glial cells after ICH. Glial cells participate in brain iron metabolism through various mechanisms; meanwhile, iron accumulation exacerbates neuroinflammation by activating inflammatory signaling pathways modulating the functions of inflammatory cells, and so on. This review aims to explore neuroinflammation from the perspective of iron metabolism, linking the complex pathophysiological changes, delving into the exploration of treatment approaches for ICH, and offering insights that could enhance clinical management strategies.
Collapse
Affiliation(s)
- Jia-Jun Ju
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
| | - Li-Hua Hang
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
- Kunshan Cancer Pain Prevention and Treatment Key Laboratory, Kunshan, China
| |
Collapse
|
20
|
Qiu L, Frazer DM, Hu M, Song R, Liu X, Qin X, Ma J, Zhou J, Tan Z, Ren F, Collins JF, Wang X. Mechanism and regulation of iron absorption throughout the life cycle. J Adv Res 2025:S2090-1232(25)00002-5. [PMID: 39814221 DOI: 10.1016/j.jare.2025.01.002] [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: 10/26/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Iron plays a crucial role through various life stages of human. Iron homeostasis is primarily regulated by iron absorption which is mediated via divalent metal-ion transporter 1 (DMT1), and iron export protein ferroportin (FPN), as there is no active pathway for iron excretion from the body. Recent studies have shown that the magnitude of iron absorption changes through various life stages to meet changing iron requirements. AIM OF REVIEW This review aims to provide an overview of recent researches on the regulation of iron absorption throughout mammalian life cycle, with the potential to reveal novel molecules and pathways at special stage of life. Such insights may pave the way for new treatments for disorders associated with aberrant iron homeostasis in the future. KEY SCIENTIFIC CONCEPTS OF REVIEW This review first summarize the mechanism and regulation of iron absorption throughout various life stages, highlighting that regulatory mechanisms have developed to precisely align iron absorption to iron requirements. In adults, iron absorption is enhanced when body is deficient of iron, conversely, iron absorption is reduced when iron demand decreases via systemic regulator Hepcidin and cellular regulation. In the elderly, age-related inflammation, hormonal changes, and chronic diseases may affect the production of Hepcidin, affecting iron absorption. In infants, intestinal iron absorption and its regulatory mechanism are different from that in adults and there might be an alternative pathway independent of DMT1 and FPN due to high iron absorption. Unique to the fetus, iron is absorbed from maternal stores for its own use through the placenta and is regulated by maternal iron status. This review also proposes directions for further studies, offering promising avenues for developing new treatments for disorders associated with aberrant iron homeostasis.
Collapse
Affiliation(s)
- Lili Qiu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - David M Frazer
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston 4029 Australia
| | - Mengxiao Hu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Rui Song
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Xiaoxue Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083 China
| | - Xiyu Qin
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Jie Ma
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Jun Zhou
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Zidi Tan
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Fazheng Ren
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083 China
| | - James F Collins
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA
| | - Xiaoyu Wang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083 China.
| |
Collapse
|
21
|
Huang J, Liu B, Zhao C, Li J, Qiu D. Degraded products generated by iron stent inhibit the vascular smooth muscle cell proliferation by downregulating AP-1. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2025; 36:7. [PMID: 39800828 PMCID: PMC11725540 DOI: 10.1007/s10856-024-06854-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
In-stent restenosis (ISR) following interventional therapy is a fatal clinical complication. Current evidence indicates that neointimal hyperplasia driven by uncontrolled proliferation of vascular smooth muscle cells (VSMC) is a major cause of restenosis. This implies that inhibiting VSMC proliferation may be an attractive approach for preventing in-stent restenosis. In our previous study, we found that the iron stent reduced the neointimal hyperplasia in an atherosclerotic artery stenosis model, and the iron corroded granules generated by the iron stent inhibited neointimal hyperplasia by suppressing the proliferation of VSMCs. However, this observation needs to be validated through in vitro experimentation. In this study, co-culture experiments and flow cytometer assays were performed to qualitatively investigate the effects of iron stent degradation on VSMCs. Moreover, the degraded products resulting generated by the iron stent were collected and used to elucidate the suppressive effect of the iron stents. The underlying mechanism was explored through molecular biology assays. The major findings are as follows: 1) The degraded iron stent inhibited the proliferation of VSMCs; 2) The degraded products of the iron stent downregulated the expression of AP-1. In summary, this study demonstrates the inhibitory effect of degraded iron products on VSMC proliferation, implying that such products have the potential to mitigate in-stent restenosis.
Collapse
Affiliation(s)
- Jiabing Huang
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, PR China
| | - Bingjian Liu
- Department of Neurology, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Chunguang Zhao
- General ICU/Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, PR China
| | - Jing Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China.
| | - Dongxu Qiu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China.
| |
Collapse
|
22
|
Kuziak A, Heczko P, Pietrzyk A, Strus M. Iron Homeostasis Dysregulation, Oro-Gastrointestinal Microbial Inflammatory Factors, and Alzheimer's Disease: A Narrative Review. Microorganisms 2025; 13:122. [PMID: 39858890 PMCID: PMC11767265 DOI: 10.3390/microorganisms13010122] [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: 12/04/2024] [Revised: 12/23/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, is a progressive neurodegenerative disorder that profoundly impacts cognitive function and the nervous system. Emerging evidence highlights the pivotal roles of iron homeostasis dysregulation and microbial inflammatory factors in the oral and gut microbiome as potential contributors to the pathogenesis of AD. Iron homeostasis disruption can result in excessive intracellular iron accumulation, promoting the generation of reactive oxygen species (ROS) and oxidative damage. Additionally, inflammatory agents produced by pathogenic bacteria may enter the body via two primary pathways: directly through the gut or indirectly via the oral cavity, entering the bloodstream and reaching the brain. This infiltration disrupts cellular homeostasis, induces neuroinflammation, and exacerbates AD-related pathology. Addressing these mechanisms through personalized treatment strategies that target the underlying causes of AD could play a critical role in preventing its onset and progression.
Collapse
Affiliation(s)
- Agata Kuziak
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, św. Łazarza 16 Street, 31-008 Cracow, Poland;
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| | - Piotr Heczko
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| | - Agata Pietrzyk
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| | - Magdalena Strus
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| |
Collapse
|
23
|
Yang H, Chen X, Chen J, Dong Y, Huang Y, Qin L, Tan J, Yi W. The pathogenesis and targeted therapies of intervertebral disc degeneration induced by cartilage endplate inflammation. Front Cell Dev Biol 2024; 12:1492870. [PMID: 39687521 PMCID: PMC11647014 DOI: 10.3389/fcell.2024.1492870] [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: 09/08/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) is the leading cause of low back pain, where degeneration and death of nucleus pulposus cells within the intervertebral disc (IVD) can be obviously revealed. This degeneration can result in an imbalance in the extracellular matrix due to the loss of proteoglycans and water content, which can further lead to catabolic and anabolic dysfunction of the IVD. Recently, the dysfunction of cartilage endplate (CEP) during aging has drawn large attention due to its essential functions in contributing nutrient exchange and maintaining IVD homeostasis. Furthermore, the inflammation and disturbed homeostasis of CEP not only accelerate the degradation of nucleus pulposus extracellular matrix, but also exacerbate IVDD by causing nucleus pulposus cell death through other pathological factors. Here in this review, we summarized the possible pathological factors and the underlying mechanisms of the CEP inflammation-induced IVDD, including exosomes degeneration, CEP calcification, ferroptosis, mechanical changes, and cell senescence. Besides, changes of miRNAs, pain-related neural reflex arc and pathways associated with CEP inflammation-induced IVDD are also reviewed. In addition, new strategies specifically designed for CEP inflammation-induced IVDD are also discussed in the last section. We hope this paper can not only offer some new insights for advancing novel strategies for treating IVDD, but also serve as a valuable reference for researchers in this field.
Collapse
Affiliation(s)
- Hantao Yang
- Department of Spine Surgery and Innovative Laboratory of Orthopedics, Shenzhen Nanshan People’s Hospital, Shenzhen, Guangdong, China
| | - Xuandu Chen
- Department of Spine Surgery and Innovative Laboratory of Orthopedics, Shenzhen Nanshan People’s Hospital, Shenzhen, Guangdong, China
| | - Jun Chen
- Orthopedic Laboratory, Orthopedic Department and Hubei Sports Medicine Center, Wuhan Fourth Hospital, Wuhan, China
| | - Yansong Dong
- Department of Spine Surgery and Innovative Laboratory of Orthopedics, Shenzhen Nanshan People’s Hospital, Shenzhen, Guangdong, China
| | - Yafang Huang
- Department of Spine Surgery and Innovative Laboratory of Orthopedics, Shenzhen Nanshan People’s Hospital, Shenzhen, Guangdong, China
- Orthopedic Laboratory, Orthopedic Department and Hubei Sports Medicine Center, Wuhan Fourth Hospital, Wuhan, China
| | - Lei Qin
- Department of Spine Surgery and Innovative Laboratory of Orthopedics, Shenzhen Nanshan People’s Hospital, Shenzhen, Guangdong, China
| | - Jie Tan
- Department of Spine Surgery and Innovative Laboratory of Orthopedics, Shenzhen Nanshan People’s Hospital, Shenzhen, Guangdong, China
- Orthopedic Laboratory, Orthopedic Department and Hubei Sports Medicine Center, Wuhan Fourth Hospital, Wuhan, China
| | - Weihong Yi
- Department of Spine Surgery and Innovative Laboratory of Orthopedics, Shenzhen Nanshan People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
24
|
Hosseinpour Mashkani SM, Bishop DP, Westerhausen MT, Adlard PA, Golzan SM. Alterations in zinc, copper, and iron levels in the retina and brain of Alzheimer's disease patients and the APP/PS1 mouse model. Metallomics 2024; 16:mfae053. [PMID: 39520546 PMCID: PMC11630249 DOI: 10.1093/mtomcs/mfae053] [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: 04/18/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Transition metals like copper (Cu), iron (Fe), and zinc (Zn) are vital for normal central nervous system function and are also linked to neurodegeneration, particularly in the onset and progression of Alzheimer's disease (AD). Their alterations in AD, identified prior to amyloid plaque aggregation, offer a unique target for staging pre-amyloid AD. However, analysing their levels in the brain is extremely challenging, necessitating the development of alternative approaches. Here, we utilized laser ablation-inductively coupled plasma-mass spectrometry and solution nebulization-inductively coupled plasma-mass spectrometry to quantitatively measure Cu, Fe, and Zn concentrations in the retina and hippocampus samples obtained from human donors (i.e. AD and healthy controls), and in the amyloid precursor protein/presenilin 1 (APP/PS1) mouse model of AD and wild-type (WT) controls, aged 9 and 18 months. Our findings revealed significantly elevated Cu, Fe, and Zn levels in the retina (*P < .05, P < .01, and P < .001) and hippocampus (*P < .05, *P < .05, and *P < .05) of human AD samples compared to healthy controls. Conversely, APP/PS1 mouse models exhibited notably lower metal levels in the same regions compared to WT mice-Cu, Fe, and Zn levels in the retina (**P < .01, *P < .05, and *P < .05) and hippocampus (**P < .01, **P < .01, and *P < .05). The contrasting metal profiles in human and mouse samples, yet similar patterns within each species' retina and brain, suggest the retina mirrors cerebral metal dyshomoeostasis in AD. Our findings lay the groundwork for staging pre-AD pathophysiology through assessment of transition metal levels in the retina.
Collapse
Affiliation(s)
- Seyed Mostafa Hosseinpour Mashkani
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - David P Bishop
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia
| | - Mika T Westerhausen
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne 3010, Australia
| | - S Mojtaba Golzan
- Vision Science Group (Orthoptics Discipline), Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
25
|
Guo Z, Huo D, Shao Y, Yang W, Wang J, Zhang Y, Xiao H, Hao B, Liao S. Novel biallelic variants in IREB2 cause an early-onset neurodegenerative disorder in a Chinese pedigree. Orphanet J Rare Dis 2024; 19:435. [PMID: 39587636 PMCID: PMC11587613 DOI: 10.1186/s13023-024-03465-7] [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: 07/25/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Cellular iron metabolism is essential for maintaining various biological processes in organisms, and this is influenced by the function of iron-responsive element-binding protein 2 (IRP2), encoded by the IREB2 gene. Since 2019, three cases of a genetic neurodegenerative syndrome resulting from compound heterozygous mutations in IREB2 have been documented, highlighting the crucial role of IRP2 in regulating iron metabolism homeostasis. This study aims to investigate the molecular basis in a single proband born to non-consanguineous healthy parents, presenting with severe psychomotor developmental abnormalities and microcytic anemia. METHODS Trio-whole exome sequencing (WES) was applied to identify the disease-causing gene in an 8-month-old male patient from China. In silico tools were used to predict the pathogenicity of the identified variants, and in vitro functional studies were performed to evaluate the molecular mechanism. RESULTS WES identified novel biallelic variants, c.1111 A > G (P.Ile371Val) and c.2477 A > T (P.Asp826Val), in the IREB2 gene, which encodes the iron metabolism-related protein, IRP2. Functional studies revealed that c.2477 A > T resulted in a significant degradation of IRP2, which led to the misregulation of intracellular ferric iron. CONCLUSIONS We report the identification of the first functional domain associated with the degradation of IRP2. The biallelic variants that affect protein degradation likely underlie the pathogenesis of the IRP2-related neurodegenerative disorder. Moreover, the use of proteasome inhibitors can potentially restore the expression of IRP2, highlighting a promising therapeutic target for patients with IRP2deficiency.
Collapse
Affiliation(s)
- Zhenglong Guo
- Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Medical Genetics Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- School of Medicine, People's Hospital of Henan University, Henan University, Zhengzhou, 450000, China.
| | - Dawei Huo
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Institute of Hematology, Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yingying Shao
- The First Affiliated Hospital of Zhengzhou University School, Zhengzhou, 450000, China
| | - Wenke Yang
- Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Medical Genetics Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
- School of Medicine, People's Hospital of Henan University, Henan University, Zhengzhou, 450000, China
| | - Jinming Wang
- Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Medical Genetics Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
- School of Medicine, People's Hospital of Henan University, Henan University, Zhengzhou, 450000, China
| | - Yuwei Zhang
- Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Medical Genetics Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
- School of Medicine, People's Hospital of Henan University, Henan University, Zhengzhou, 450000, China
| | - Hai Xiao
- Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Medical Genetics Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
- School of Medicine, People's Hospital of Henan University, Henan University, Zhengzhou, 450000, China
| | - Bingtao Hao
- Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Medical Genetics Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450000, China.
| | - Shixiu Liao
- Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Medical Genetics Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- School of Medicine, People's Hospital of Henan University, Henan University, Zhengzhou, 450000, China.
| |
Collapse
|
26
|
Wang H, Gao J, Wen L, Huang K, Liu H, Zeng L, Zeng Z, Liu Y, Mo Z. Ion channels in acinar cells in acute pancreatitis: crosstalk of calcium, iron, and copper signals. Front Immunol 2024; 15:1444272. [PMID: 39606246 PMCID: PMC11599217 DOI: 10.3389/fimmu.2024.1444272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
The initial stages of acute pancreatitis (AP) are characterized by a significant event - acinar ductal metaplasia (ADM). This process is a crucial feature of both acute and chronic pancreatitis, serving as the first step in the development of pancreatic cancer. Ion channels are integral transmembrane proteins that play a pivotal role in numerous biological processes by modulating ion flux. In many diseases, the expression and activity of ion channels are often dysregulated. Metal ions, including calcium ions (Ca2+), ferrous ions (Fe2+), and Copper ions (Cu2+), assume a distinctive role in cellular metabolism. These ions possess specific biological properties relevant to cellular function. However, the interactions among these ions exacerbate the imbalance within the intracellular environment, resulting in cellular damage and influencing the progression of AP. A more in-depth investigation into the mechanisms by which these ions interact with acinar cells is essential for elucidating AP's pathogenesis and identifying novel therapeutic strategies. Currently, treatment for AP primarily focuses on pain relief, complications prevention, and prognosis improvement. There are limited specific treatments targeting acinous cell dedifferentiation or ion imbalance. This study aims to investigate potential therapeutic strategies by examining ion crosstalk within acinar cells in the context of acute pancreatitis.
Collapse
Affiliation(s)
- Hanli Wang
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Jianhua Gao
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Lingling Wen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kejun Huang
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Huixian Liu
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Linsheng Zeng
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Zhongyi Zeng
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yuxiang Liu
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
27
|
Kanome Y, Ohtomo S, Nakatsu M, Kohno M, Fukui K. Effect of cerium oxide on iron metabolism in mice. J Clin Biochem Nutr 2024; 75:190-196. [PMID: 39583972 PMCID: PMC11579851 DOI: 10.3164/jcbn.24-38] [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: 03/04/2024] [Accepted: 04/27/2024] [Indexed: 11/26/2024] Open
Abstract
The use of metal nanoparticles such as cerium oxide nanoparticles (nanoceria) in living organisms is attracting increasing attention. We administered nanoceria to chronic kidney disease model rats, including a 5/6 nephrectomy model and adenine administration model rats, and reported high phosphorus adsorption capacity and renal function improvement effects of nanoceria. However, the iron ion concentration in the serum fluctuated significantly after administration. Therefore, we investigated changes in proteins related to iron metabolism following administration of nanoceria to normal mice without chronic kidney disease over different periods of time. Nanoceria were administered to 10-week-old C57BL/6 mice for 4 or 12 weeks. Another group was administrated lanthanum carbonate, which is currently used as a phosphorus adsorbent. The amount of iron in the serum and the concentration of transferrin in the liver were significantly increased following nanoceria administration, and the amount of iron in the liver was significantly decreased. There were no changes in serum hepcidin, ferroportin, cholesterol, or low-density lipoprotein levels. These results indicate that nanoceria administration can affect iron metabolism in mice. Although the detailed mechanism remains unknown, caution is warranted when considering biological utilization in the future.
Collapse
Affiliation(s)
- Yuki Kanome
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| | - Shunki Ohtomo
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| | - Masaharu Nakatsu
- applause Company Limited, Biko building 4F, Shinkawa 2-24-2, Chuo-ku, Tokyo 104-0033, Japan
| | - Masahiro Kohno
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| | - Koji Fukui
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| |
Collapse
|
28
|
Wu L, Lin H, Li S, Huang Y, Sun Y, Shu S, Luo T, Liang T, Lai W, Rao J, Hu Z, Peng H. Macrophage iron dyshomeostasis promotes aging-related renal fibrosis. Aging Cell 2024; 23:e14275. [PMID: 39016438 PMCID: PMC11561705 DOI: 10.1111/acel.14275] [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: 12/22/2023] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
Renal aging, marked by the accumulation of senescent cells and chronic low-grade inflammation, leads to renal interstitial fibrosis and impaired function. In this study, we investigate the role of macrophages, a key regulator of inflammation, in renal aging by analyzing kidney single-cell RNA sequencing data of C57BL/6J mice from 8 weeks to 24 months. Our findings elucidate the dynamic changes in the proportion of kidney cell types during renal aging and reveal that increased macrophage infiltration contributes to chronic low-grade inflammation, with these macrophages exhibiting senescence and activation of ferroptosis signaling. CellChat analysis indicates enhanced communications between macrophages and tubular cells during aging. Suppressing ferroptosis alleviates macrophage-mediated tubular partial epithelial-mesenchymal transition in vitro, thereby mitigating the expression of fibrosis-related genes. Using SCENIC analysis, we infer Stat1 as a key age-related transcription factor promoting iron dyshomeostasis and ferroptosis in macrophages by regulating the expression of Pcbp1, an iron chaperone protein that inhibits ferroptosis. Furthermore, through virtual screening and molecular docking from a library of anti-aging compounds, we construct a docking model targeting Pcbp1, which indicates that the natural small molecule compound Rutin can suppress macrophage senescence and ferroptosis by preserving Pcbp1. In summary, our study underscores the crucial role of macrophage iron dyshomeostasis and ferroptosis in renal aging. Our results also suggest Pcbp1 as an intervention target in aging-related renal fibrosis and highlight Rutin as a potential therapeutic agent in mitigating age-related renal chronic low-grade inflammation and fibrosis.
Collapse
Affiliation(s)
- Lingzhi Wu
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Hongchun Lin
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shaomin Li
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuebo Huang
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuxiang Sun
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shuangshuang Shu
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ting Luo
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Tiantian Liang
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Weiyan Lai
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jialing Rao
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhaoyong Hu
- Nephrology Division, Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - Hui Peng
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhouChina
| |
Collapse
|
29
|
Yin M, Liu Z, Sun Z, Qu X, Chen Z, Diao Y, Cheng Y, Shen S, Wang X, Cai Z, Lu B, Tan S, Wang Y, Zhao X, Chen F. Biomimetic Scaffolds Regulating the Iron Homeostasis for Remolding Infected Osteogenic Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407251. [PMID: 39373362 PMCID: PMC11600272 DOI: 10.1002/advs.202407251] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/12/2024] [Indexed: 10/08/2024]
Abstract
The treatment of infected bone defects (IBDs) needs simultaneous elimination of infection and acceleration of bone regeneration. One mechanism that hinders the regeneration of IBDs is the iron competition between pathogens and host cells, leading to an iron deficient microenvironment that impairs the innate immune responses. In this work, an in situ modification strategy is proposed for printing iron-active multifunctional scaffolds with iron homeostasis regulation ability for treating IBDs. As a proof-of-concept, ultralong hydroxyapatite (HA) nanowires are modified through in situ growth of a layer of iron gallate (FeGA) followed by incorporation in the poly(lactic-co-glycolic acid) (PLGA) matrix to print biomimetic PLGA based composite scaffolds containing FeGA modified HA nanowires (FeGA-HA@PLGA). The photothermal effect of FeGA endows the scaffolds with excellent antibacterial activity. The released iron ions from the FeGA-HA@PLGA help restore the iron homeostasis microenvironment, thereby promoting anti-inflammatory, angiogenesis and osteogenic differentiation. The transcriptomic analysis shows that FeGA-HA@PLGA scaffolds exert anti-inflammatory and pro-osteogenic differentiation by activating NF-κB, MAPK and PI3K-AKT signaling pathways. Animal experiments confirm the excellent bone repair performance of FeGA-HA@PLGA scaffolds for IBDs, suggesting the promising prospect of iron homeostasis regulation therapy in future clinical applications.
Collapse
Affiliation(s)
- Mengting Yin
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Zhiqing Liu
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Zhongyi Sun
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital & School of StomatologyFudan UniversityShanghai201102P. R. China
- Suzhou First People's HospitalSchool of MedicineAnhui University of Science and TechnologyAnhui232001P.R. China
| | - Xinyu Qu
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Ziyan Chen
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Yuying Diao
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital & School of StomatologyFudan UniversityShanghai201102P. R. China
| | - Yuxuan Cheng
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Sisi Shen
- Department of Plastic and Reconstructive SurgeryShanghai Key Laboratory of Tissue EngineeringShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Xiansong Wang
- Department of Plastic and Reconstructive SurgeryShanghai Key Laboratory of Tissue EngineeringShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Zhuyun Cai
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Bingqiang Lu
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Shuo Tan
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Yan Wang
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital & School of StomatologyFudan UniversityShanghai201102P. R. China
| | - Xinyu Zhao
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Feng Chen
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital & School of StomatologyFudan UniversityShanghai201102P. R. China
- Suzhou First People's HospitalSchool of MedicineAnhui University of Science and TechnologyAnhui232001P.R. China
| |
Collapse
|
30
|
Chen Y, Meng Z, Li Y, Liu S, Hu P, Luo E. Advanced glycation end products and reactive oxygen species: uncovering the potential role of ferroptosis in diabetic complications. Mol Med 2024; 30:141. [PMID: 39251935 PMCID: PMC11385660 DOI: 10.1186/s10020-024-00905-9] [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: 04/16/2024] [Accepted: 08/19/2024] [Indexed: 09/11/2024] Open
Abstract
Advanced glycation end products (AGEs) are a diverse range of compounds that are formed when free amino groups of proteins, lipids, and nucleic acids are carbonylated by reactive carbonyl species or glycosylated by reducing sugars. Hyperglycemia in patients with diabetes can cause an overabundance of AGEs. Excess AGEs are generally acknowledged as major contributing factors to the development of diabetic complications because of their ability to break down the extracellular matrix directly and initiate intracellular signaling pathways by binding to the receptor for advanced glycation end products (RAGE). Inflammation and oxidative stress are the two most well-defined pathophysiological states induced by the AGE-RAGE interaction. In addition to oxidative stress, AGEs can also inhibit antioxidative systems and disturb iron homeostasis, all of which may induce ferroptosis. Ferroptosis is a newly identified contributor to diabetic complications. This review outlines the formation of AGEs in individuals with diabetes, explores the oxidative damage resulting from downstream reactions of the AGE-RAGE axis, and proposes a novel connection between AGEs and the ferroptosis pathway. This study introduces the concept of a vicious cycle involving AGEs, oxidative stress, and ferroptosis in the development of diabetic complications.
Collapse
Affiliation(s)
- Yanchi Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zihan Meng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yong Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Pei Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
31
|
Wu Y, Xiao M, Chen J, Tao Y, Chen A, Lin H, Xu Y, Li L, Jia H, Xue Y, Jia Y, Zheng Z. Association of dietary iron intake with diabetic kidney disease among individuals with diabetes. Endocrine 2024; 85:1154-1161. [PMID: 38758293 DOI: 10.1007/s12020-024-03819-y] [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: 12/11/2023] [Accepted: 04/02/2024] [Indexed: 05/18/2024]
Abstract
PURPOSE The current study investigated the correlation between dietary iron intake and diabetic kidney disease among diabetic adults. METHODS This cross-sectional study enrolled 8118 participants who suffered from diabetes from the National Health and Nutrition Examination Survey (NHANES) 1999-2018. Dietary iron intake was obtained from 24 h recall interviews, and diabetic kidney disease was defined as eGFR < 60 mL/min per 1.73 m2 or albumin creatinine ratio (ACR) ≥ 30 mg/g. Three weighted logistic regression models were utilized to investigate odd ratio (OR) and 95% CIs for diabetic kidney disease. Stratified analyses were performed by gender, age, BMI, HbA1c, hypertension status, and smoking status, and diabetes types. RESULTS Among 8118 participants (51.6% male, mean age 61.3 years), 40.7% of participants suffered from diabetic kidney disease. With the adjustment of potential covariates, we found that ≥ 12.59 mg of dietary iron was related to a lower risk of diabetic kidney disease (OR = 0.78, 95% CI: 0.63 to 0.96; OR = 0.79, 95% CI: 0.63 to 0.98). In stratified analyses, higher iron intake was negatively related to diabetic kidney disease, especially among those who were male, < 60 years, those with hypertension, those with HbA1c < 7.0%, and those who were ex-smokers. The result remained robust in sensitivity analyses. CONCLUSION We found that ≥ 12.59 mg of dietary iron is associated with a lower risk of diabetic kidney disease, especially in those who were male, younger, heavier weight, have better blood sugar control, and those who were ex-smokers.
Collapse
Affiliation(s)
- Yichuan Wu
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Manlu Xiao
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaqi Chen
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Tao
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Aomiao Chen
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Huanjia Lin
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Ying Xu
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- De Feng Academy, Southern Medical University, Guangzhou, China
| | - Linna Li
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongxia Jia
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yaoming Xue
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yijie Jia
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- De Feng Academy, Southern Medical University, Guangzhou, China.
| | - Zongji Zheng
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- De Feng Academy, Southern Medical University, Guangzhou, China.
| |
Collapse
|
32
|
Zeidan RS, Yoon HS, Yang JJ, Sobh A, Braithwaite D, Mankowski R, Leeuwenburgh C, Anton S. Iron and cancer: overview of the evidence from population-based studies. Front Oncol 2024; 14:1393195. [PMID: 39246326 PMCID: PMC11377248 DOI: 10.3389/fonc.2024.1393195] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/23/2024] [Indexed: 09/10/2024] Open
Abstract
Iron is an essential nutrient required for various physiological processes in the body. However, iron imbalance can potentially contribute to initiating and promoting cancer development. Epidemiological studies have investigated the relationship between dietary iron intake and the risk of different types of cancer, yet, not all studies have consistently shown a significant association between dietary iron and cancer risk. Also, studies have shown different effects of dietary heme and non-heme iron intake on cancer risk. While some epidemiological studies suggest a possible link between high dietary iron (mainly heme-iron) intake and increased cancer risk, the evidence remains inconsistent. Moreover, multiple iron biomarkers, which can mirror physiological iron status, have demonstrated varied correlations with the risk of cancer, contingent upon the specific biomarker analyzed and the type of cancer being investigated. Here, we have investigated the current evidence on the potential relationship between dietary iron intake on one hand, and iron biomarkers on the other hand, with the risk of developing different types of cancer, including breast, prostate, lung, pancreatic, colon, colorectal, and liver cancers. Further research is warranted to better understand the complex relationship between dietary iron, physiological iron and cancer development. Future research should account for factors that affect and interact with dietary iron and physiological iron levels, such as genetic susceptibility, overall diet quality, and lifestyle habits.
Collapse
Affiliation(s)
- Rola S Zeidan
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Hyung-Suk Yoon
- Cancer Control and Population Science Division, University of Florida Health Cancer Center, Gainesville, FL, United States
- Division of Hematology and Oncology, University of Florida Health Cancer Center, Gainesville, FL, United States
| | - Jae Jeong Yang
- Cancer Control and Population Science Division, University of Florida Health Cancer Center, Gainesville, FL, United States
- Division of Hematology and Oncology, University of Florida Health Cancer Center, Gainesville, FL, United States
| | - Amin Sobh
- Division of Hematology and Oncology, University of Florida Health Cancer Center, Gainesville, FL, United States
| | - Dejana Braithwaite
- Cancer Control and Population Science Division, University of Florida Health Cancer Center, Gainesville, FL, United States
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Robert Mankowski
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Christian Leeuwenburgh
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Stephen Anton
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
33
|
Agostini F, Sgalletta B, Bisaglia M. Iron Dyshomeostasis in Neurodegeneration with Brain Iron Accumulation (NBIA): Is It the Cause or the Effect? Cells 2024; 13:1376. [PMID: 39195264 PMCID: PMC11352641 DOI: 10.3390/cells13161376] [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: 07/22/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Iron is an essential metal ion implicated in several cellular processes. However, the reactive nature of iron renders this metal ion potentially dangerous for cells, and its levels need to be tightly controlled. Alterations in the intracellular concentration of iron are associated with different neuropathological conditions, including neurodegeneration with brain iron accumulation (NBIA). As the name suggests, NBIA encompasses a class of rare and still poorly investigated neurodegenerative disorders characterized by an abnormal accumulation of iron in the brain. NBIA is mostly a genetic pathology, and to date, 10 genes have been linked to familial forms of NBIA. In the present review, after the description of the principal mechanisms implicated in iron homeostasis, we summarize the research data concerning the pathological mechanisms underlying the genetic forms of NBIA and discuss the potential involvement of iron in such processes. The picture that emerges is that, while iron overload can contribute to the pathogenesis of NBIA, it does not seem to be the causal factor in most forms of the pathology. The onset of these pathologies is rather caused by a combination of processes involving the interplay between lipid metabolism, mitochondrial functions, and autophagic activity, eventually leading to iron dyshomeostasis.
Collapse
Affiliation(s)
- Francesco Agostini
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Bibiana Sgalletta
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Marco Bisaglia
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, 35121 Padova, Italy
| |
Collapse
|
34
|
Zhu L, Cai X, Cai Y, Shi W, Jiang Y. Unveiling the causal relationship between circulating levels of micronutrients and risk of facial aging: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e39118. [PMID: 39093739 PMCID: PMC11296447 DOI: 10.1097/md.0000000000039118] [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: 05/20/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024] Open
Abstract
Facial aging involves a continuous sequence of complex, interrelated events that impact numerous facial tissues. The aim of the study was to elucidate the casual relationship between circulating micronutrients and risk of facial aging. A two-sample Mendelian randomization analysis was performed using genetic data from genome-wide association studies. The inverse-variance weighted method is used for causal effect estimation, and additional tools such as Mendelian randomization-Egger, weighted median, simple mode, and weighted mode were used to refine the analysis. We conducted an in-depth examination of the correlation between several micronutrient blood levels and the risk of facial aging, and identified 3 key micronutrients (selenium, carotene, and iron) that may have a significant impact on skin health. Inverse-variance weighted results indicate that selenium levels were positively correlated with the risk of facial aging (odds ratio [OR] 1.005, P = .027), while a negative causal effect of carotene (OR 0.979, P = .024) and iron (OR 0.976, P = .009) on age-related facial alterations was observed. This study offers a new and insightful perspective on the current understanding of antiaging strategies, particularly the importance of appropriate consumption of essential micronutrients to maintain healthy skin condition.
Collapse
Affiliation(s)
- Lijiao Zhu
- Medical Aesthetic Department, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Xueyao Cai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuchen Cai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjun Shi
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiyuan Jiang
- Medical Aesthetic Department, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| |
Collapse
|
35
|
Nishizawa H, Matsumoto M, Yamanaka M, Irikura R, Nakajima K, Tada K, Nakayama Y, Konishi M, Itoh N, Funayama R, Nakayama K, Igarashi K. BACH1 inhibits senescence, obesity, and short lifespan by ferroptotic FGF21 secretion. Cell Rep 2024; 43:114403. [PMID: 38943639 DOI: 10.1016/j.celrep.2024.114403] [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: 10/17/2023] [Revised: 04/14/2024] [Accepted: 06/11/2024] [Indexed: 07/01/2024] Open
Abstract
Ferroptosis is a type of regulated cell death characterized by iron-dependent lipid peroxidation. A model cell system is constructed to induce ferroptosis by re-expressing the transcription factor BACH1, a potent ferroptosis inducer, in immortalized mouse embryonic fibroblasts (iMEFs). The transfer of the culture supernatant from ferroptotic iMEFs activates the proliferation of hepatoma cells and other fibroblasts and suppresses cellular senescence-like features. The BACH1-dependent secretion of the longevity factor FGF21 is increased in ferroptotic iMEFs. The anti-senescent effects of the culture supernatant from these iMEFs are abrogated by Fgf21 knockout. BACH1 activates the transcription of Fgf21 by promoting ferroptotic stress and increases FGF21 protein expression by suppressing its autophagic degradation through transcriptional Sqstm1 and Lamp2 repression. The BACH1-induced ferroptotic FGF21 secretion suppresses obesity in high-fat diet-fed mice and the short lifespan of progeria mice. The inhibition of these aging-related phenotypes can be physiologically significant regarding ferroptosis.
Collapse
Affiliation(s)
- Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan.
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Gladstone Institute of Neurological Disease, Gladstone Institute, San Francisco, CA 94158, USA
| | - Riko Irikura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Kazuma Nakajima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Keisuke Tada
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Yoshiaki Nakayama
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Hyogo 658-8558, Japan
| | - Morichika Konishi
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Hyogo 658-8558, Japan
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo, Kyoto 606-8501, Japan
| | - Ryo Funayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Keiko Nakayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan.
| |
Collapse
|
36
|
Fontes A, Jauch AT, Sailer J, Engler J, Azul AM, Zischka H. Metabolic Derangement of Essential Transition Metals and Potential Antioxidant Therapies. Int J Mol Sci 2024; 25:7880. [PMID: 39063122 PMCID: PMC11277342 DOI: 10.3390/ijms25147880] [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: 05/19/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Essential transition metals have key roles in oxygen transport, neurotransmitter synthesis, nucleic acid repair, cellular structure maintenance and stability, oxidative phosphorylation, and metabolism. The balance between metal deficiency and excess is typically ensured by several extracellular and intracellular mechanisms involved in uptake, distribution, and excretion. However, provoked by either intrinsic or extrinsic factors, excess iron, zinc, copper, or manganese can lead to cellular damage upon chronic or acute exposure, frequently attributed to oxidative stress. Intracellularly, mitochondria are the organelles that require the tightest control concerning reactive oxygen species production, which inevitably leaves them to be one of the most vulnerable targets of metal toxicity. Current therapies to counteract metal overload are focused on chelators, which often cause secondary effects decreasing patients' quality of life. New therapeutic options based on synthetic or natural antioxidants have proven positive effects against metal intoxication. In this review, we briefly address the cellular metabolism of transition metals, consequences of their overload, and current therapies, followed by their potential role in inducing oxidative stress and remedies thereof.
Collapse
Affiliation(s)
- Adriana Fontes
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, D-85764 Neuherberg, Germany;
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Adrian T. Jauch
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| | - Judith Sailer
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| | - Jonas Engler
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| | - Anabela Marisa Azul
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, D-85764 Neuherberg, Germany;
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| |
Collapse
|
37
|
Guo C, Peng J, Cheng P, Yang C, Gong S, Zhang L, Zhang T, Peng J. Mechanistic elucidation of ferroptosis and ferritinophagy: implications for advancing our understanding of arthritis. Front Physiol 2024; 15:1290234. [PMID: 39022306 PMCID: PMC11251907 DOI: 10.3389/fphys.2024.1290234] [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: 09/07/2023] [Accepted: 02/23/2024] [Indexed: 07/20/2024] Open
Abstract
In recent years, the emerging phenomenon of ferroptosis has garnered significant attention as a distinctive mode of programmed cell death. Distinguished by its reliance on iron and dependence on reactive oxygen species (ROS), ferroptosis has emerged as a subject of extensive investigation. Mechanistically, this intricate process involves perturbations in iron homeostasis, dampening of system Xc-activity, morphological dynamics within mitochondria, and the onset of lipid peroxidation. Additionally, the concomitant phenomenon of ferritinophagy, the autophagic degradation of ferritin, assumes a pivotal role by facilitating the liberation of iron ions from ferritin, thereby advancing the progression of ferroptosis. This discussion thoroughly examines the detailed cell structures and basic processes behind ferroptosis and ferritinophagy. Moreover, it scrutinizes the intricate web of regulators that orchestrate these processes and examines their intricate interplay within the context of joint disorders. Against the backdrop of an annual increase in cases of osteoarthritis, rheumatoid arthritis, and gout, these narrative sheds light on the intriguing crossroads of pathophysiology by dissecting the intricate interrelationships between joint diseases, ferroptosis, and ferritinophagy. The newfound insights contribute fresh perspectives and promising therapeutic avenues, potentially revolutionizing the landscape of joint disease management.
Collapse
Affiliation(s)
- Caopei Guo
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Jiaze Peng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Piaotao Cheng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Chengbing Yang
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Shouhang Gong
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Lin Zhang
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiachen Peng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
- Department of Burn and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, China
| |
Collapse
|
38
|
Chen Y, Zhang J, Tian Y, Xu X, Wang B, Huang Z, Lou S, Kang J, Zhang N, Weng J, Liang Y, Ma W. Iron accumulation in ovarian microenvironment damages the local redox balance and oocyte quality in aging mice. Redox Biol 2024; 73:103195. [PMID: 38781731 PMCID: PMC11145558 DOI: 10.1016/j.redox.2024.103195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Accumulating oxidative damage is a primary driver of ovarian reserve decline along with aging. However, the mechanism behind the imbalance in reactive oxygen species (ROS) is not yet fully understood. Here we investigated changes in iron metabolism and its relationship with ROS disorder in aging ovaries of mice. We found increased iron content in aging ovaries and oocytes, along with abnormal expression of iron metabolic proteins, including heme oxygenase 1 (HO-1), ferritin heavy chain (FTH), ferritin light chain (FTL), mitochondrial ferritin (FTMT), divalent metal transporter 1 (DMT1), ferroportin1(FPN1), iron regulatory proteins (IRP1 and IRP2) and transferrin receptor 1 (TFR1). Notably, aging oocytes exhibited enhanced ferritinophagy and mitophagy, and consistently, there was an increase in cytosolic Fe2+, elevated lipid peroxidation, mitochondrial dysfunction, and augmented lysosome activity. Additionally, the ovarian expression of p53, p21, p16 and microtubule-associated protein tau (Tau) were also found to be upregulated. These alterations could be phenocopied with in vitro Fe2+ administration in oocytes from 2-month-old mice but were alleviated by deferoxamine (DFO). In vivo application of DFO improved ovarian iron metabolism and redox status in 12-month-old mice, and corrected the alterations in cytosolic Fe2+, ferritinophagy and mitophagy, as well as related degenerative changes in oocytes. Thereby in the whole, DFO delayed the decline in ovarian reserve and significantly increased the number of superovulated oocytes with reduced fragmentation and aneuploidy. Together, our findings suggest that aging-related disturbance in ovarian iron homeostasis contributes to excessive ROS production and that iron chelation may improve ovarian redox status, and efficiently delay the decline in ovarian reserve and oocyte quality in aging mice. These data propose a novel intervention strategy for preserving the ovarian reserve function in elderly women.
Collapse
Affiliation(s)
- Ye Chen
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jiaqi Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ying Tian
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiangning Xu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Bicheng Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ziqi Huang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Shuo Lou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jingyi Kang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ningning Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jing Weng
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yuanjing Liang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wei Ma
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
39
|
Pellowski D, Heinze T, Tuchtenhagen M, Müller SM, Meyer S, Maares M, Gerbracht C, Wernicke C, Haase H, Kipp AP, Grune T, Pfeiffer AFH, Mai K, Schwerdtle T. Fostering healthy aging through selective nutrition: A long-term comparison of two dietary patterns and their holistic impact on mineral status in middle-aged individuals-A randomized controlled intervention trial in Germany. J Trace Elem Med Biol 2024; 84:127462. [PMID: 38701651 DOI: 10.1016/j.jtemb.2024.127462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/06/2024] [Accepted: 04/19/2024] [Indexed: 05/05/2024]
Abstract
Aging is associated with a decline in physiological functions and an increased risk of age-related diseases, emphasizing the importance of identifying dietary strategies for healthy aging. Minerals play a crucial role in maintaining optimal health during aging, making them relevant targets for investigation. Therefore, we aimed to analyze the effect of different dietary pattern on mineral status in the elderly. We included 502 individuals aged 50-80 years in a 36-month randomized controlled trial (RCT) (NutriAct study). This article focuses on the results within the two-year intervention period. NutriAct is not a mineral-modulating-targeted intervention study, rather examining nutrition in the context of healthy aging in general. However, mineral status might be affected in an incidental manner. Participants were assigned to either NutriAct dietary pattern (proportionate intake of total energy consumption (%E) of 35-45 %E carbohydrates, 35-40 %E fats, and 15-25 %E protein) or the German Nutrition Society (DGE) dietary pattern (proportionate intake of total energy consumption (%E) of 55 %E carbohydrates, 30 %E fats, and 15 %E protein), differing in the composition of macronutrients. Data from 368 participants regarding dietary intake (energy, calcium, magnesium, iron, and zinc) and serum mineral concentrations of calcium, magnesium, iron, copper, zinc, selenium, iodine, and manganese, free zinc, and selenoprotein P were analyzed at baseline, as well as after 12 and 24 months to gain comprehensive insight into the characteristics of the mineral status. Additionally, inflammatory status - sensitive to changes in mineral status - was assessed by measurement of C-reactive protein and interleukin-6. At baseline, inadequate dietary mineral intake and low serum concentrations of zinc and selenium were observed in both dietary patterns. Throughout two years, serum zinc concentrations decreased, while an increase of serum selenium, manganese and magnesium concentrations was observable, likely influenced by both dietary interventions. No significant changes were observed for serum calcium, iron, copper, or iodine concentrations. In conclusion, long-term dietary interventions can influence serum mineral concentrations in a middle-aged population. Our findings provide valuable insights into the associations between dietary habits, mineral status, and disease, contributing to dietary strategies for healthy aging.
Collapse
Affiliation(s)
- Denny Pellowski
- Trace-Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Nuthetal, 14558, Germany; Institute of Nutritional Science, Department Food Chemistry, University of Potsdam, Potsdam 14469, Germany; NutriAct Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, 14558, Germany
| | - Tom Heinze
- Trace-Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Nuthetal, 14558, Germany; Institute of Nutritional Science, Department Food Chemistry, University of Potsdam, Potsdam 14469, Germany
| | - Max Tuchtenhagen
- Trace-Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Nuthetal, 14558, Germany; Institute of Nutritional Science, Department Food Chemistry, University of Potsdam, Potsdam 14469, Germany
| | - Sandra M Müller
- Institute of Nutritional Science, Department Food Chemistry, University of Potsdam, Potsdam 14469, Germany; NutriAct Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, 14558, Germany
| | - Sören Meyer
- Institute of Nutritional Science, Department Food Chemistry, University of Potsdam, Potsdam 14469, Germany
| | - Maria Maares
- Trace-Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Nuthetal, 14558, Germany; Department of Food Chemistry and Toxicology, Technische Universität Berlin, Berlin 13355, Germany
| | - Christiana Gerbracht
- Human Study Center, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, 14558, Germany
| | - Charlotte Wernicke
- NutriAct Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, 14558, Germany; Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin 10117, Germany
| | - Hajo Haase
- Trace-Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Nuthetal, 14558, Germany; Department of Food Chemistry and Toxicology, Technische Universität Berlin, Berlin 13355, Germany
| | - Anna P Kipp
- Trace-Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Nuthetal, 14558, Germany; Department of Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena 07743, Germany
| | - Tilman Grune
- Trace-Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Nuthetal, 14558, Germany; NutriAct Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, 14558, Germany; Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, 14558, Germany; Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Andreas F H Pfeiffer
- NutriAct Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, 14558, Germany; Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin 10117, Germany
| | - Knut Mai
- NutriAct Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, 14558, Germany; Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin 10117, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, 85764, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin 10115, Germany; Department of Human Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, Nuthetal, 14558, Germany
| | - Tanja Schwerdtle
- Trace-Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Nuthetal, 14558, Germany; NutriAct Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, 14558, Germany; German Federal Institute for Risk Assessment (BfR), Berlin 10589, Germany.
| |
Collapse
|
40
|
Hu Y, Huang Y, Zong L, Lin J, Liu X, Ning S. Emerging roles of ferroptosis in pulmonary fibrosis: current perspectives, opportunities and challenges. Cell Death Discov 2024; 10:301. [PMID: 38914560 PMCID: PMC11196712 DOI: 10.1038/s41420-024-02078-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial lung disorder characterized by abnormal myofibroblast activation, accumulation of extracellular matrix (ECM), and thickening of fibrotic alveolar walls, resulting in deteriorated lung function. PF is initiated by dysregulated wound healing processes triggered by factors such as excessive inflammation, oxidative stress, and coronavirus disease (COVID-19). Despite advancements in understanding the disease's pathogenesis, effective preventive and therapeutic interventions are currently lacking. Ferroptosis, an iron-dependent regulated cell death (RCD) mechanism involving lipid peroxidation and glutathione (GSH) depletion, exhibits unique features distinct from other RCD forms (e.g., apoptosis, necrosis, and pyroptosis). Imbalance between reactive oxygen species (ROS) production and detoxification leads to ferroptosis, causing cellular dysfunction through lipid peroxidation, protein modifications, and DNA damage. Emerging evidence points to the crucial role of ferroptosis in PF progression, driving macrophage polarization, fibroblast proliferation, and ECM deposition, ultimately contributing to alveolar cell death and lung tissue scarring. This review provides a comprehensive overview of the latest findings on the involvement and signaling mechanisms of ferroptosis in PF pathogenesis, emphasizing potential novel anti-fibrotic therapeutic approaches targeting ferroptosis for PF management.
Collapse
Affiliation(s)
- Yixiang Hu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China
| | - Ying Huang
- Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, China
| | - Lijuan Zong
- Department of Rehabilitation Medicine, Zhongda Hospital of Southeast University, Nanjing, 210096, China
| | - Jiaxin Lin
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Xiang Liu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
41
|
Xu W, Jiang T, Ding L, Jiang Y, Zhang L, Xia T, Xin H. Bajitianwan formula extract ameliorates bone loss induced by iron overload via activating RAGE/PI3K/AKT pathway based on network pharmacology and transcriptomic analysis. J Nat Med 2024; 78:488-504. [PMID: 38530577 DOI: 10.1007/s11418-024-01779-1] [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: 11/06/2023] [Accepted: 01/03/2024] [Indexed: 03/28/2024]
Abstract
Osteoporosis (OP) is closely related to iron overload. Bajitianwan (BJTW) is a traditional Chinese medicine formulation used for treating senile diseases such as dementia and osteoporosis. Modern pharmacological researches have found that BJTW has beneficial effect on bone loss and memory impairment in aging rats. This paper aimed to explore the role and mechanism of BJTW in ameliorating iron overload-induced bone loss. Furthermore, BJTW effectively improved the bone micro-structure of the femur in mice, and altered bone metabolism biomarkers alkaline phosphatase (ALP) and osteocalcin (OCN) in serum, as well as oxidative indexes superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GR) glutathione (GSH) and malondialdehyde (MDA) in liver. As for network pharmacology, 73 components collected from BJTW regulated 99 common targets merged in the BJTW and OP. The results of RNA-seq indicated that there were 418 potential targets in BJTW low dose group (BJTW-L) and 347 potential targets in BJTW high dose group (BJTW-H). Intriguingly, both PI3K-AKT signaling pathway and the AGEs-RAGE signaling pathway were contained in the KEGG pathways enrichment results of network pharmacology and transcriptomics, which were considered as the potential mechanism. Additionally, we verified that BJTW regulated the expression of related proteins in RAGE/PI3K-AKT pathways in MC3T3-E1 cells. In summary, BJTW has potent effect on protecting against iron overload-induced OP, and its mechanism may be related to the activation of the RAGE/PI3K-AKT signaling pathways.
Collapse
Affiliation(s)
- Weifan Xu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Tao Jiang
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Luying Ding
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yiping Jiang
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Lichao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Tianshuang Xia
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
| | - Hailiang Xin
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
42
|
Zheng Q, Wang F, Nie C, Zhang K, Sun Y, Al-Ansi W, Wu Q, Wang L, Du J, Li Y. Elevating the significance of legume intake: A novel strategy to counter aging-related mitochondrial dysfunction and physical decline. Compr Rev Food Sci Food Saf 2024; 23:e13342. [PMID: 38634173 DOI: 10.1111/1541-4337.13342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024]
Abstract
Mitochondrial dysfunction increasingly becomes a target for promoting healthy aging and longevity. The dysfunction of mitochondria with age ultimately leads to a decline in physical functions. Among them, biogenesis dysfunction and the imbalances in the metabolism of reactive oxygen species and mitochondria as signaling organelles in the aging process have aroused our attention. Dietary intervention in mitochondrial dysfunction and physical decline during aging processes is essential, and greater attention should be directed toward healthful legume intake. Legumes are constantly under investigation for their nutritional and bioactive properties, and their consumption may yield antiaging and mitochondria-protecting benefits. This review summarizes mitochondrial dysfunction with age, discusses the benefits of legumes on mitochondrial function, and introduces the potential role of legumes in managing aging-related physical decline. Additionally, it reveals the benefits of legume intake for the elderly and offers a viable approach to developing legume-based functional food.
Collapse
Affiliation(s)
- Qingwei Zheng
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Feijie Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Chenzhipeng Nie
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Kuiliang Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yujie Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Waleed Al-Ansi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qiming Wu
- Nutrilite Health Institute, Shanghai, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jun Du
- Nutrilite Health Institute, Shanghai, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
43
|
Zeidan RS, Martenson M, Tamargo JA, McLaren C, Ezzati A, Lin Y, Yang JJ, Yoon HS, McElroy T, Collins JF, Leeuwenburgh C, Mankowski RT, Anton S. Iron homeostasis in older adults: balancing nutritional requirements and health risks. J Nutr Health Aging 2024; 28:100212. [PMID: 38489995 DOI: 10.1016/j.jnha.2024.100212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
Iron plays a crucial role in many physiological processes, including oxygen transport, bioenergetics, and immune function. Iron is assimilated from food and also recycled from senescent red blood cells. Iron exists in two dietary forms: heme (animal based) and non-heme (mostly plant based). The body uses iron for metabolic purposes, and stores the excess mainly in splenic and hepatic macrophages. Physiologically, iron excretion in humans is inefficient and not highly regulated, so regulation of intestinal absorption maintains iron homeostasis. Iron losses occur at a steady rate via turnover of the intestinal epithelium, blood loss, and exfoliation of dead skin cells, but overall iron homeostasis is tightly controlled at cellular and systemic levels. Aging can have a profound impact on iron homeostasis and induce a dyshomeostasis where iron deficiency or overload (sometimes both simultaneously) can occur, potentially leading to several disorders and pathologies. To maintain physiologically balanced iron levels, reduce risk of disease, and promote healthy aging, it is advisable for older adults to follow recommended daily intake guidelines and periodically assess iron levels. Clinicians can evaluate body iron status using different techniques but selecting an assessment method primarily depends on the condition being examined. This review provides a comprehensive overview of the forms, sources, and metabolism of dietary iron, associated disorders of iron dyshomeostasis, assessment of iron levels in older adults, and nutritional guidelines and strategies to maintain iron balance in older adults.
Collapse
Affiliation(s)
- Rola S Zeidan
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Matthew Martenson
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Javier A Tamargo
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Christian McLaren
- Department of Clinical and Health Psychology, College of Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Armin Ezzati
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Food, Nutrition, Dietetics and Health, Kansas State University, Manhattan, KS, USA
| | - Yi Lin
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jae Jeong Yang
- UF Health Cancer Center, Gainesville, FL, USA; Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Hyung-Suk Yoon
- UF Health Cancer Center, Gainesville, FL, USA; Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Taylor McElroy
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - James F Collins
- Department of Food Science & Human Nutrition, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | - Christiaan Leeuwenburgh
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Robert T Mankowski
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Stephen Anton
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Clinical and Health Psychology, College of Health and Health Professions, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
44
|
Chen H, Zhang Z, Wang Y, Ma A, Li L, Zhao G. Iron status and sarcopenia-related traits: a bi-directional Mendelian randomization study. Sci Rep 2024; 14:9179. [PMID: 38649459 PMCID: PMC11035655 DOI: 10.1038/s41598-024-60059-w] [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: 12/13/2023] [Accepted: 04/18/2024] [Indexed: 04/25/2024] Open
Abstract
Although serum iron status and sarcopenia are closely linked, the presence of comprehensive evidence to establish a causal relationship between them remains insufficient. The objective of this study is to employ Mendelian randomization techniques to clarify the association between serum iron status and sarcopenia. We conducted a bi-directional Mendelian randomization (MR) analysis to investigate the potential causal relationship between iron status and sarcopenia. MR analyses were performed using inverse variance weighted (IVW), MR-Egger, and weighted median methods. Additionally, sensitivity analyses were conducted to verify the reliability of the causal association results. Then, we harvested a combination of SNPs as an integrated proxy for iron status to perform a MVMR analysis based on IVW MVMR model. UVMR analyses based on IVW method identified causal effect of ferritin on appendicular lean mass (ALM, β = - 0.051, 95% CI - 0.072, - 0.031, p = 7.325 × 10-07). Sensitivity analyses did not detect pleiotropic effects or result fluctuation by outlying SNPs in the effect estimates of four iron status on sarcopenia-related traits. After adjusting for PA, the analysis still revealed that each standard deviation higher genetically predicted ferritin was associated with lower ALM (β = - 0.054, 95% CI - 0.092, - 0.015, p = 0.006). Further, MVMR analyses determined a predominant role of ferritin (β = - 0.068, 95% CI - 0.12, - 0.017, p = 9.658 × 10-03) in the associations of iron status with ALM. Our study revealed a causal association between serum iron status and sarcopenia, with ferritin playing a key role in this relationship. These findings contribute to our understanding of the complex interplay between iron metabolism and muscle health.
Collapse
Affiliation(s)
- Honggu Chen
- Department of Orthopedics, the Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, Jiangsu Province, People's Republic of China
| | - Ziyi Zhang
- Department of Orthopedics, the Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, Jiangsu Province, People's Republic of China
| | - Yizhe Wang
- School of Medicine of Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Anpei Ma
- Department of Orthopedics, Yancheng First People's Hospital, Yancheng, 224000, Jiangsu Province, People's Republic of China
| | - Lingbo Li
- Department of Internal Medicine, Peking Union Medical College Hospital, Beijing, 100730, Beijing, People's Republic of China
| | - Guoyang Zhao
- Department of Orthopedics, the Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, Jiangsu Province, People's Republic of China.
| |
Collapse
|
45
|
Jia H, Chang Y, Chen Y, Chen X, Zhang H, Hua X, Xu M, Sheng Y, Zhang N, Cui H, Han L, Zhang J, Fu X, Song J. A single-cell atlas of lung homeostasis reveals dynamic changes during development and aging. Commun Biol 2024; 7:427. [PMID: 38589700 PMCID: PMC11001898 DOI: 10.1038/s42003-024-06111-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 03/26/2024] [Indexed: 04/10/2024] Open
Abstract
Aging is a global challenge, marked in the lungs by function decline and structural disorders, which affects the health of the elderly population. To explore anti-aging strategies, we develop a dynamic atlas covering 45 cell types in human lungs, spanning from embryonic development to aging. We aim to apply the discoveries of lung's development to address aging-related issues. We observe that both epithelial and immune cells undergo a process of acquisition and loss of essential function as they transition from development to aging. During aging, we identify cellular phenotypic alternations that result in reduced pulmonary compliance and compromised immune homeostasis. Furthermore, we find a distinctive expression pattern of the ferritin light chain (FTL) gene, which increases during development but decreases in various types of lung cells during the aging process.
Collapse
Affiliation(s)
- Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yulin Chen
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xiao Chen
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hang Zhang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengda Xu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yixuan Sheng
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningning Zhang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Cui
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Han
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of General Surgery, Yanan Hospital, Kunming Medical University, Kunming, China
| | - Jian Zhang
- Thoracic Surgery Department, the third affiliated hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China.
| | - Xiaodong Fu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
46
|
Chen Y, Wu Z, Li S, Chen Q, Wang L, Qi X, Tian C, Yang M. Mapping the Research of Ferroptosis in Parkinson's Disease from 2013 to 2023: A Scientometric Review. Drug Des Devel Ther 2024; 18:1053-1081. [PMID: 38585257 PMCID: PMC10999190 DOI: 10.2147/dddt.s458026] [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: 01/04/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
Methods Related studies on PD and ferroptosis were searched in Web of Science Core Collection (WOSCC) from inception to 2023. VOSviewer, CiteSpace, RStudio, and Scimago Graphica were employed as bibliometric analysis tools to generate network maps about the collaborations between authors, countries, and institutions and to visualize the co-occurrence and trends of co-cited references and keywords. Results A total of 160 original articles and reviews related to PD and ferroptosis were retrieved, produced by from 958 authors from 162 institutions. Devos David was the most prolific author, with 9 articles. China and the University of Melbourne had leading positions in publication volume with 84 and 12 publications, respectively. Current hot topics focus on excavating potential new targets for treating PD based on ferroptosis by gaining insight into specific molecular mechanisms, including iron metabolism disorders, lipid peroxidation, and imbalanced antioxidant regulation. Clinical studies aimed at treating PD by targeting ferroptosis remain in their preliminary stages. Conclusion A continued increase was shown in the literature within the related field over the past decade. The current study suggested active collaborations among authors, countries, and institutions. Research into the pathogenesis and treatment of PD based on ferroptosis has remained a prominent topic in the field in recent years, indicating that ferroptosis-targeted therapy is a potential approach to halting the progression of PD.
Collapse
Affiliation(s)
- Yingfan Chen
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Zhenhui Wu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, People’s Republic of China
| | - Shaodan Li
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Qi Chen
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Liang Wang
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Xiaorong Qi
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
| | - Chujiao Tian
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
| | - Minghui Yang
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
47
|
Rosell-Díaz M, Santos-González E, Motger-Albertí A, Gallardo-Nuell L, Arnoriaga-Rodríguez M, Coll-Martínez C, Ramió-Torrentà L, Garre-Olmo J, Puig J, Ramos R, Mayneris-Perxachs J, Fernández-Real JM. Lower serum ferritin levels are associated with worse cognitive performance in aging. J Nutr Health Aging 2024; 28:100190. [PMID: 38368845 DOI: 10.1016/j.jnha.2024.100190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/20/2024]
Abstract
OBJECTIVES Iron is important for neurogenesis, synaptic development, and neurotransmitter synthesis. Serum ferritin (SF) is a reliable marker for assessing iron stores. Therefore, we evaluated the cognitive function associated with SF levels. We also assessed brain iron content using R2* Magnetic Resonance Imaging (MRI) and its association with SF levels. DESIGN Data from three cross-sectional observational studies were used. Aging Imageomics (n = 1030) was conducted on aged subjects. Health Imageomics (n = 971) and IR0NMET (n = 175) were conducted in middle-aged subjects. SETTING AND PARTICIPANTS Participants were enrolled at Dr. Josep Trueta University Hospital facilities. The three cohorts included a total of 2176 subjects (mean age, 52 years; 48% men). MEASUREMENTS SF levels were measured by standard laboratory methods. Total Digits Span (TDS), and Phonemic Verbal Fluency (PVF) were used to assess executive function. Language function was assessed by semantic verbal fluency (SVF), attention by the Symbol Digit Modalities Test, and memory by the Memory Binding Tests - Total Free Recall and Total Delayed Free Recall. MRI was used to assess the iron content of the brain by R2*. RESULTS In subjects aged 65 years or older, SF levels were associated with increased TDS (β = 0.003, p = 0.02), PVF (β = 0.004, p = 0.01), and SVF (β = 0.004, p = 0.002) scores. After stratification by sex, these findings were significant only in men, where SF was associated with increased TDS (β = 0.003, p = 0.01), PVF (β = 0.004, p = 0.03), and SVF (β = 0.004, p = 0.009) scores. In middle-aged subjects, SF was also associated with increased SVF scores (β = 0.005, p = 0.011). Lastly, in men, SF levels were negatively associated with R2*, a surrogate marker of brain iron content, in both the left frontal inferior opercular area (r = -0.41, p = 0.005) and the right frontal inferior opercular area (r = -0.44, p = 0.002). CONCLUSIONS SF is significantly and positively associated with cognition. In older people with low SF levels, iron supplementation may be a promising therapy to improve cognition.
Collapse
Affiliation(s)
- Marisel Rosell-Díaz
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain; CIBER Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Elena Santos-González
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain; CIBER Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Anna Motger-Albertí
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain; CIBER Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Laura Gallardo-Nuell
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain; CIBER Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - María Arnoriaga-Rodríguez
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain; CIBER Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Clàudia Coll-Martínez
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Dr. Josep Trueta Hospital, Neurodegeneration and Neuroinflammation Research Group, (IDIBGI-CERCA), Girona, Spain; Research Group on Health, Gender, and Aging, Girona Biomedical Research Institute (IDIBGI-CERCA) and University of Girona, Girona, Spain
| | - Lluís Ramió-Torrentà
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Dr. Josep Trueta Hospital, Neurodegeneration and Neuroinflammation Research Group, (IDIBGI-CERCA), Girona, Spain; Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
| | - Josep Garre-Olmo
- Research Group on Health, Gender, and Aging, Girona Biomedical Research Institute (IDIBGI-CERCA) and University of Girona, Girona, Spain; Serra-Hunter Professor, Department of Nursing, University of Girona, Spain
| | - Josep Puig
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain; Radiology Department CDI, Hospital Clinic of Barcelona, Barcelona, Spain; Medical Imaging, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain
| | - Rafael Ramos
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain; Vascular Health Research Group of Girona (ISV-Girona), Jordi Gol Institute for Primary Care Research (Institut Universitari per a la Recerca en Atenció Primària Jordi Gol I Gorina -IDIAPJGol), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud-RICAPPS- ISCIII, Spain; Girona Biomedical Research Institute (IDIBGI-CERCA), Dr. Josep Trueta University Hospital, Catalonia, Spain
| | - Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain; CIBER Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain.
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain; CIBER Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain; Girona Biomedical Research Institute (IDIBGI-CERCA), Dr. Josep Trueta University Hospital, Catalonia, Spain.
| |
Collapse
|
48
|
Wang W, Ma Z, Feng X, Ren J, Sun S, Shao Y, Zhang W, Yang X, Zhang J, Jing X. TfR1 mediated iron metabolism dysfunction as a potential therapeutic target for osteoarthritis. Arthritis Res Ther 2024; 26:71. [PMID: 38493104 PMCID: PMC10943767 DOI: 10.1186/s13075-024-03304-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024] Open
Abstract
OBJECTIVE Transferrin receptor-1 (TfR1) plays important roles in controlling cellular iron levels, but its role in OA pathology is unknown. Herein we aim to investigate the role of TfR1 in OA progression and its underlying mechanisms. METHODS TfR1 expression in cartilage during OA development were examined both in vivo and in vitro. Then IL-1β was used to induce chondrocytes degeneration in vitro and TfR1 siRNA was used for observing the effect of TfR1 in modulating iron homeostasis, mitochondrial function and degrading enzymes expression. Also the inhibitor of TfR1 was exploited to analyze the protective effect of TfR1 inhibition in vivo. RESULTS TfR1 is elevated in OA cartilage and contributes to OA inflammation condition. Excess iron not only results in oxidative stress damage and sensitizes chondrocytes to ferroptosis, but also triggers c-GAS/STING-mediated inflammation by promoting mitochondrial destruction and the release of mtDNA. Silencing TfR1 using TfR1 siRNA not only reduced iron content in chondrocytes and inhibited oxidative stress, but also facilitated the mitophagy process and suppressed mtDNA/cGAS/STING-mediated inflammation. Importantly, we also found that Ferstatin II, a novel and selective TfR1 inhibitor, could substantially suppress TfR1 activity both in vivo and in vitro and ameliorated cartilage degeneration. CONCLUSION Our work demonstrates that TfR1 mediated iron influx plays important roles in chondrocytes degeneration and OA pathogenesis, suggesting that maintaining iron homeostasis through the targeting of TfR1 may represent a novel therapeutic strategy for the treatment of OA.
Collapse
Affiliation(s)
- Wenchao Wang
- Department of Spine Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Zhenkai Ma
- Department of Neurosurgery, Binzhou People's Hospital, Binzhou, 256600, China
| | - Xuemin Feng
- Department of Endocrinology, Binzhou People's Hospital, Binzhou, 256600, China
| | - Jiabin Ren
- Department of Spine Surgery, Binzhou Medical University Hospital, Binzhou, 256600, China
| | - Shengyao Sun
- Shandong First Medical University & Shandong Academy of Medical Sciences, No.6699 Qingdao Road, Jinan, 250117, China
| | - Yuandong Shao
- Department of Spine Surgery, Binzhou People's Hospital, Binzhou, 256600, China
| | - Weimin Zhang
- Department of Spine Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Xiaoxia Yang
- Department of Spine Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Jiaming Zhang
- Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China.
| | - Xingzhi Jing
- Department of Spine Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250000, Shandong, China.
| |
Collapse
|
49
|
Xu Y, Zeng Q, Zhang A. Assessing the mechanisms and adjunctive therapy for arsenic-induced liver injury in rats. ENVIRONMENTAL TOXICOLOGY 2024; 39:1197-1209. [PMID: 37902164 DOI: 10.1002/tox.24008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 07/03/2023] [Accepted: 10/09/2023] [Indexed: 10/31/2023]
Abstract
Environmental arsenic exposure is a significant global public health concern. Previous studies have demonstrated the association between arsenic-induced liver injury and oxidative stress as well as ferroptosis. However, the knowledge of the interactions among these mechanisms remains limited. Moreover, there is a lack of research on potential therapeutic interventions for liver injury resulting from arsenic exposure. To address these limitations, we established a rat model with liver injury caused by arsenic exposure and investigated the impact of the nuclear factor E2-related factor 2 (Nrf2)/glutathione peroxidase 4 (GPx4) signaling pathway and ferroptosis on arsenic-induced liver injury. Our findings revealed that arsenic increased Nrf2 expression and decreased GPx4 expression in the rat liver. This was accompanied by a substantial generation of reactive oxygen species and disruption of the antioxidant defense system, ultimately promoting liver injury through ferroptosis. Subsequently, we conducted intervention experiments using Rosa roxburghii Tratt (RRT) in rats exposed to arsenic. The results showed that the detrimental effects mentioned earlier were partially alleviated following RRT intervention. This study offers preliminary evidence that persistent activation of Nrf2 by arsenic triggers an adaptive antioxidant response, leading to liver injury through the promotion of ferroptosis. Additionally, we discovered that RRT inhibits Nrf2-mediated adaptive antioxidant responses by reducing hepatic ferroptosis, thereby mitigating liver injury caused by arsenic exposure in rats. Our study contributes to a deeper understanding of the molecular mechanisms underlying liver injury resulting from arsenic exposure. Furthermore, our findings may facilitate the identification of a potential edible and medicinal plant extracts that could be utilized to develop a more effective adjunctive treatment approach.
Collapse
Affiliation(s)
- Yuyan Xu
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, China
| | - Qibing Zeng
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, China
| | - Aihua Zhang
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, China
| |
Collapse
|
50
|
Punziano C, Trombetti S, Cesaro E, Grosso M, Faraonio R. Antioxidant Systems as Modulators of Ferroptosis: Focus on Transcription Factors. Antioxidants (Basel) 2024; 13:298. [PMID: 38539832 PMCID: PMC10967371 DOI: 10.3390/antiox13030298] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 10/28/2024] Open
Abstract
Ferroptosis is a type of programmed cell death that differs from apoptosis, autophagy, and necrosis and is related to several physio-pathological processes, including tumorigenesis, neurodegeneration, senescence, blood diseases, kidney disorders, and ischemia-reperfusion injuries. Ferroptosis is linked to iron accumulation, eliciting dysfunction of antioxidant systems, which favor the production of lipid peroxides, cell membrane damage, and ultimately, cell death. Thus, signaling pathways evoking ferroptosis are strongly associated with those protecting cells against iron excess and/or lipid-derived ROS. Here, we discuss the interaction between the metabolic pathways of ferroptosis and antioxidant systems, with a particular focus on transcription factors implicated in the regulation of ferroptosis, either as triggers of lipid peroxidation or as ferroptosis antioxidant defense pathways.
Collapse
Affiliation(s)
- Carolina Punziano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| | - Silvia Trombetti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy
| | - Elena Cesaro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| | - Raffaella Faraonio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| |
Collapse
|