1
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [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: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
2
|
Catapano A, Cimmino F, Petrella L, Pizzella A, D'Angelo M, Ambrosio K, Marino F, Sabbatini A, Petrelli M, Paolini B, Lucchin L, Cavaliere G, Cristino L, Crispino M, Trinchese G, Mollica MP. Iron metabolism and ferroptosis in health and diseases: The crucial role of mitochondria in metabolically active tissues. J Nutr Biochem 2025; 140:109888. [PMID: 40057002 DOI: 10.1016/j.jnutbio.2025.109888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/30/2025]
Abstract
Iron is essential in various physiological processes, but its accumulation leads to oxidative stress and cell damage, thus iron homeostasis has to be tightly regulated. Ferroptosis is an iron-dependent non-apoptotic regulated cell death characterized by iron overload and reactive oxygen species accumulation. Mitochondria are organelles playing a crucial role in iron metabolism and involved in ferroptosis. MitoNEET, a protein of mitochondrial outer membrane, is a key element in this process. Ferroptosis, altering iron levels in several metabolically active organs, is linked to several non-communicable diseases. For example, iron overload in the liver leads to hepatic fibrosis and cirrhosis, accelerating non-alcholic fatty liver diseases progression, in the muscle cells contributes to oxidative damage leading to sarcopenia, and in the brain is associated to neurodegeneration. The aim of this review is to investigate the intricate balance of iron regulation focusing on the role of mitochondria and oxidative stress, and analyzing the ferroptosis implications in health and disease.
Collapse
Affiliation(s)
- Angela Catapano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy; Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Lidia Petrella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Katia Ambrosio
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Francesca Marino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Annarita Sabbatini
- Dietetic and Clinical Nutrition Unit, IEO European Institute of Oncology IRCSS, Milan, Italy
| | - Massimiliano Petrelli
- Department of Clinical and Molecular Sciences, Clinic of Endocrinology and Metabolic Diseases, Università Politecnica delle Marche, Ancona, Italy
| | - Barbara Paolini
- Department of Innovation, experimentation and clinical research, Unit of dietetics and clinical nutrition, S. Maria Alle Scotte Hospital, University of Siena, Siena, Italy
| | - Lucio Lucchin
- Dietetics and Clinical Nutrition, Bolzano Health District, Bolzano, Italy
| | - Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | | | | |
Collapse
|
3
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [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: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
4
|
Jiang C, Yan Y, Long T, Xu J, Chang C, Kang M, Wang X, Chen Y, Qiu J. Ferroptosis: a potential therapeutic target in cardio-cerebrovascular diseases. Mol Cell Biochem 2025:10.1007/s11010-025-05262-7. [PMID: 40148662 DOI: 10.1007/s11010-025-05262-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Cardio-cerebrovascular diseases (CCVDs) are the leading cause of global mortality, yet effective treatment options remain limited. Ferroptosis, a novel form of regulated cell death, has emerged as a critical player in various CCVDs, including atherosclerosis, myocardial infarction, ischemia-reperfusion injury, cardiomyopathy, and ischemic/hemorrhagic strokes. This review highlights the core mechanisms of ferroptosis, its pathological implications in CCVDs, and the therapeutic potential of targeting this process. Additionally, it explores the role of Chinese herbal medicines (CHMs) in mitigating ferroptosis, offering novel therapeutic strategies for CCVDs management. Ferroptosis is regulated by several key pathways. The GPX4-GSH-System Xc- axis is central to ferroptosis execution, involving GPX4 using GSH to neutralize lipid peroxides, with system Xc- being crucial for GSH synthesis. The NAD(P)H/FSP1/CoQ10 axis involves FSP1 regenerating CoQ10 via NAD(P)H, inhibiting lipid peroxidation independently of GPX4. Lipid peroxidation, driven by PUFAs and enzymes like ACSL4 and LPCAT3, and iron metabolism, regulated by proteins like TfR1 and ferritin, are also crucial for ferroptosis. Inhibiting ferroptosis shows promise in managing CCVDs. In atherosclerosis, ferroptosis inhibitors reduce iron accumulation and lipid peroxidation. In myocardial infarction, inhibitors protect cardiomyocytes by preserving GPX4 and SLC7A11 levels. In ischemia-reperfusion injury, targeting ferroptosis reduces myocardial and cerebral damage. In diabetic cardiomyopathy, Nrf2 activators alleviate oxidative stress and iron metabolism irregularities. CHMs offer natural compounds that mitigate ferroptosis. They possess antioxidant properties, chelate iron, and modulate signaling pathways like Nrf2 and AMPK. For example, Salvia miltiorrhiza and Astragalus membranaceus reduce oxidative stress, while some CHMs chelate iron, reducing its availability for ferroptosis. In conclusion, ferroptosis plays a pivotal role in CCVDs, and targeting it offers novel therapeutic avenues. CHMs show promise in reducing ferroptosis and improving patient outcomes. Future research should explore combination therapies and further elucidate the molecular interactions in ferroptosis.
Collapse
Affiliation(s)
- Chenlong Jiang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Yang Yan
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Tianlin Long
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Jiawei Xu
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Cuicui Chang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Meili Kang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Xuanqi Wang
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| | - Yuhua Chen
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China.
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China.
- School of Life and Health Science, Hainan University, No. 58 People's Avenue, Haikou, 570100, Hainan, China.
| | - Junlin Qiu
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| |
Collapse
|
5
|
Liu C, Pan J, Bao Q. Ferroptosis in senescence and age-related diseases: pathogenic mechanisms and potential intervention targets. Mol Biol Rep 2025; 52:238. [PMID: 39960579 DOI: 10.1007/s11033-025-10338-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/04/2025] [Indexed: 05/09/2025]
Abstract
As the global population continues to age, the prevalence of age-related diseases is increasing, significantly influencing social and economic development, the stability of social security systems, and progress in medical technology. Ferroptosis, a recently discovered form of programmed cell death driven by iron-dependent lipid peroxidation, has emerged as a key area of research. Studies have revealed a strong association between ferroptosis and senescence. In this article, we systematically summarize the molecular mechanisms and associated signaling pathways underlying ferroptosis, emphasizing its pivotal role in the onset and progression of age-related diseases. By providing new perspectives, we aim to advance understanding of the pathogenesis of age-related diseases and guide the development of effective intervention strategies.
Collapse
Affiliation(s)
- Chang Liu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Pan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Qi Bao
- Zhejiang University School of Medicine, Hangzhou, China.
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
6
|
Suárez-Ortegón MF, McLachlan S, Fernández-Real JM, Wilson JF, Wild SH. Both low and high body iron stores relate to metabolic syndrome in postmenopausal women: Findings from the VIKING Health Study-Shetland (VIKING I). Eur J Clin Invest 2024; 54:e14312. [PMID: 39239983 DOI: 10.1111/eci.14312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND There are conflicting results among studies on the association between serum ferritin (SF) and metabolic syndrome (MetS), and by groups of sex/menopausal status. To date, there are no studies on British populations. The SF-MetS association might be U/J-shaped. We evaluated whether SF was independently associated with MetS (harmonized definition) in people from Shetland, Scotland. METHODS We analysed cross-sectional data from the Viking Health Study-Shetland (589 premenopausal women [PreMW], 625 postmenopausal women [PostW] and 832 men). Logistic regressions using two approaches, one with the lowest sex and menopausal status-specific ferritin quartile (Q) as the reference and other using the middle two quartiles combined (2-3) as the reference, were conducted to estimate the SF-MetS association. The shape of the association was verified via cubic spline analyses. The associations were adjusted for age, inflammatory and hepatic injury markers, alcohol intake, smoking and BMI. RESULTS Prevalence of MetS was 18.3%. Among PostMW both low and high SF were associated with MetS (fully adjusted odds ratios [95% confidence interval] compared to the middle two quartiles combined were: 1.99 [1.17-3.38] p =.011 for Q1 and 2.10 [1.27-3.49] p =.004 for Q4) This U-shaped pattern was confirmed in the cubic spline analysis in PostMW with a ferritin range of 15-200 ug/L. In men, a positive association between ferritin quartiles with Q1 as the reference, did not remain significant after adjustment for BMI. CONCLUSION Extreme quartiles of iron status were positively associated with MetS in PostMW, while no SF-MetS associations were found in men or PreMW. The ferritin-MetS association pattern differs between populations and U/J-shaped associations may exist.
Collapse
Affiliation(s)
| | | | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Institut d'Investigació Biomèdica de Girona (IDIBGI-CERCA), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, Edinburgh, UK
- Centre for Genomic Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Sarah H Wild
- Usher Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
7
|
Haase T, Ludwig A, Stach A, Mohtashamdolatshahi A, Hauptmann R, Mundhenk L, Kratz H, Metzkow S, Kader A, Freise C, Mueller S, Stolzenburg N, Radon P, Liebl M, Wiekhorst F, Hamm B, Taupitz M, Schnorr J. Repeated Injection of Very Small Superparamagnetic Iron Oxide Particles (VSOPs) in Murine Atherosclerosis: A Safety Study. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:773. [PMID: 38727367 PMCID: PMC11085881 DOI: 10.3390/nano14090773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
Citrate-coated electrostatically stabilized very small superparamagnetic iron oxide particles (VSOPs) have been successfully tested as magnetic resonance angiography (MRA) contrast agents and are promising tools for molecular imaging of atherosclerosis. Their repeated use in the background of pre-existing hyperlipidemia and atherosclerosis has not yet been studied. This study aimed to investigate the effect of multiple intravenous injections of VSOPs in atherosclerotic mice. Taurine-formulated VSOPs (VSOP-T) were repeatedly intravenously injected at 100 µmol Fe/kg in apolipoprotein E-deficient (ApoE KO) mice with diet-induced atherosclerosis. Angiographic imaging was carried out by in vivo MRI. Magnetic particle spectrometry was used to detect tissue VSOP content, and tissue iron content was quantified photometrically. Pathological changes in organs, atherosclerotic plaque development, and expression of hepatic iron-related proteins were evaluated. VSOP-T enabled the angiographic imaging of heart and blood vessels with a blood half-life of one hour. Repeated intravenous injection led to VSOP deposition and iron accumulation in the liver and spleen without affecting liver and spleen pathology, expression of hepatic iron metabolism proteins, serum lipids, or atherosclerotic lesion formation. Repeated injections of VSOP-T doses sufficient for MRA analyses had no significant effects on plaque burden, steatohepatitis, and iron homeostasis in atherosclerotic mice. These findings underscore the safety of VSOP-T and support its further development as a contrast agent and molecular imaging tool.
Collapse
Affiliation(s)
- Tobias Haase
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Antje Ludwig
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10117 Berlin, Germany
| | - Anke Stach
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Azadeh Mohtashamdolatshahi
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Ralf Hauptmann
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Lars Mundhenk
- Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, 14163 Berlin, Germany;
| | - Harald Kratz
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Susanne Metzkow
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Avan Kader
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Christian Freise
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Susanne Mueller
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
- Charité 3R|Replace, Reduce, Refine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Nicola Stolzenburg
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Patricia Radon
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany; (P.R.); (M.L.); (F.W.)
| | - Maik Liebl
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany; (P.R.); (M.L.); (F.W.)
| | - Frank Wiekhorst
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany; (P.R.); (M.L.); (F.W.)
| | - Bernd Hamm
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Matthias Taupitz
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Jörg Schnorr
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
8
|
Yuan XM, Sultana N, Ghosh-Laskar M, Li W. Elevated Hepcidin Expression in Human Carotid Atheroma: Sex-Specific Differences and Associations with Plaque Vulnerability. Int J Mol Sci 2024; 25:1706. [PMID: 38338987 PMCID: PMC10855936 DOI: 10.3390/ijms25031706] [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: 12/17/2023] [Revised: 01/19/2024] [Accepted: 01/27/2024] [Indexed: 02/12/2024] Open
Abstract
Hepcidin is upregulated by increased body iron stores and inflammatory cytokines. It is associated with cardiovascular events, arterial stiffness, and increased iron accumulation in human atheroma with hemorrhage. However, it is unknown whether the expression of hepcidin in human carotid plaques is related to plaque severity and whether hepcidin expression differs between men and women. Carotid samples from 58 patients (38 males and 20 females) were immunostained with hepcidin, macrophages, ferritin, and transferrin receptor. Immunocytochemistry of hepcidin was performed on THP-1 macrophages exposed to iron or 7betahydroxycholesterol. Hepcidin expression significantly increases with the progression of human atherosclerotic plaques. Plaques of male patients have significantly higher levels of hepcidin. Expressions of hepcidin are significantly correlated with the accumulation of CD68-positive macrophages and transferrin receptor 1 (TfR1) and apoptosis. In vitro, hepcidin is significantly increased in macrophages exposed to iron and moderately increased following 7-oxysterol treatment. In the cultured cells, suppression of hepcidin protected against macrophage cell death, lysosomal membrane permeabilization, and oxidative stress. Hepcidin may play a crucial role in the development and progression of atherosclerosis. The differential expression of hepcidin in male and female patients and its significant correlations with plaque severity, highlight the potential of hepcidin as a biomarker for risk stratification and therapeutic targeting in atherosclerosis.
Collapse
Affiliation(s)
- Xi-Ming Yuan
- Occupational and Environmental Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, 581 85 Linköping, Sweden;
| | - Nargis Sultana
- Laboratory Medicine, Linköping University Hospital, 581 85 Linköping, Sweden;
| | | | - Wei Li
- Obstetrics and Gynaecology in Linköping, Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| |
Collapse
|
9
|
Arumugam S, Suyambulingam A. Association Between Serum Ferritin and the Duration of Type 2 Diabetes Mellitus in a Tertiary Care Hospital in Chennai. Cureus 2024; 16:e53117. [PMID: 38420099 PMCID: PMC10900170 DOI: 10.7759/cureus.53117] [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: 12/15/2023] [Accepted: 01/28/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Hyperinsulinemia has been linked to increased ferritin production and iron absorption in type 2 diabetes mellitus, ultimately leading to increased iron storage. Glucose intolerance is intimately linked to this issue. Increased oxidative stress from iron decreases insulin's ability to be taken into cells and used for energy. Researchers suggest that increased iron levels in the body play a role in the emergence of insulin resistance, glucose intolerance, and vascular repercussions associated with diabetes. OBJECTIVE The aim of this study is to assess the levels of serum ferritin and fasting plasma glucose in both diabetic and nondiabetic individuals while establishing a relationship between the two. Exploring the connection between serum ferritin levels and the duration of diabetes mellitus in individuals diagnosed with diabetes is our objective. METHODOLOGY In this study, 80 men diagnosed with type 2 diabetes mellitus were included, and they were compared with 70 male volunteers who were in good health. We took blood samples while the subjects fasted, and we analyzed the plasma glucose and serum ferritin levels. RESULTS In the diabetic group, there were notably higher levels of serum ferritin and fasting plasma glucose compared to the nondiabetic subjects. Furthermore, a correlation was observed between the duration of diabetes among participants with diabetes and elevated serum ferritin levels. CONCLUSION The findings suggest that low-grade inflammation and increased body iron stores are positively related to hyperglycemia in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Sankar Arumugam
- General Medicine, Sree Balaji Medical College and Hospital, Chennai, IND
| | | |
Collapse
|
10
|
Magnussen LV, Helskov Jørgensen L, Glintborg D, Andersen MS. Hepcidin Reduction during Testosterone Therapy in Men with Type 2 Diabetes: A Randomized, Double-Blinded, Placebo-Controlled Study. Biomedicines 2023; 11:3184. [PMID: 38137405 PMCID: PMC10740671 DOI: 10.3390/biomedicines11123184] [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: 10/09/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
High hepcidin is linked to low-grade inflammation and lower iron levels. The consequences of testosterone replacement therapy (TRT) on inflammation and the risk of cardiovascular disease (CVD) are undetermined. We investigate the effect of TRT on the inflammatory cardiovascular risk markers hepcidin-iron, fibroblast growth factor 23 (FGF23)-phosphate-klotho, and calprotectin pathways. METHODS A randomized, placebo-controlled, double-blinded study at an academic tertiary-care medical center. Interventions were testosterone gel (TRT, n = 20) or placebo gel (n = 19) for 24 weeks. We included 39 men (50-70 years) with type 2 diabetes (T2D) on metformin monotherapy with bioavailable testosterone levels <7.3 nmol/L. Body composition was assessed with DXA- and MRI-scans; the main study outcomes were serum hepcidin-iron, FGF23, phosphate, klotho, and calprotectin. RESULTS Hepcidin levels decreased during TRT (β = -9.5 ng/mL, p < 0.001), lean body mass (β = 1.9 kg, p = 0.001) increased, and total fat mass (β = -1.3 kg, p = 0.009) decreased compared to placebo. Delta hepcidin was not associated with changes in lean body mass or fat mass. Iron and the pathways of FGF23-phosphate-klotho and calprotectin were unchanged during TRT. CONCLUSIONS During TRT, the reduction in hepcidin was not associated with circulating iron levels, lean body mass, or fat mass; these findings suggested a direct anti-inflammatory effect of TRT and no indirect effect mediated through these factors.
Collapse
Affiliation(s)
- Line Velling Magnussen
- Department of Endocrinology and Metabolism, Odense University Hospital, 5000 Odense, Denmark; (D.G.); (M.S.A.)
| | - Louise Helskov Jørgensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark;
| | - Dorte Glintborg
- Department of Endocrinology and Metabolism, Odense University Hospital, 5000 Odense, Denmark; (D.G.); (M.S.A.)
| | - Marianne Skovsager Andersen
- Department of Endocrinology and Metabolism, Odense University Hospital, 5000 Odense, Denmark; (D.G.); (M.S.A.)
| |
Collapse
|
11
|
Halliwell B, Watt F, Minqin R. Iron and atherosclerosis: Lessons learned from rabbits relevant to human disease. Free Radic Biol Med 2023; 209:165-170. [PMID: 37852545 DOI: 10.1016/j.freeradbiomed.2023.10.383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023]
Abstract
The role of iron in promoting atherosclerosis, and hence the cardiovascular, neurodegenerative and other diseases that result from atherosclerosis, has been fiercely controversial. Many studies have been carried out on various rodent models of atherosclerosis, especially on apoE-knockout (apoE-/-) mice, which develop atherosclerosis more readily than normal mice. These apoE-/- mouse studies generally support a role for iron in atherosclerosis development, although there are conflicting results. The purpose of the current article is to describe studies on another animal model that is not genetically manipulated; New Zealand White (NZW) rabbits fed a high-cholesterol diet. This may be a better model than the apoE-/- mice for human atherosclerosis, although it has been given much less attention. Studies on NZW rabbits support the view that iron promotes atherosclerosis, although some uncertainties remain, which need to be resolved by further experimentation.
Collapse
Affiliation(s)
- Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Research Programme, National University of Singapore, Centre for Life Sciences, #05-01A, 28 Medical Drive, 117456, Singapore.
| | - Frank Watt
- Department of Physics, National University of Singapore, Faculty of Science, 2 Science Drive 3, Blk S12, Level 2, 117551, Singapore.
| | - Ren Minqin
- Department of Physics, National University of Singapore, Faculty of Science, 2 Science Drive 3, Blk S12, Level 2, 117551, Singapore.
| |
Collapse
|
12
|
Guo Q, Qian C, Qian ZM. Iron metabolism and atherosclerosis. Trends Endocrinol Metab 2023:S1043-2760(23)00090-5. [PMID: 37210298 DOI: 10.1016/j.tem.2023.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/22/2023]
Abstract
Despite several decades of study, whether iron is involved in the development of atherosclerosis remains a controversial and unresolved issue. Here, we focus on the up-to-date advances in studies on role of iron in atherosclerosis and discuss possible reasons why patients with hereditary hemochromatosis (HH) do not show any increased incidence of atherosclerosis. In addition, we analyze conflicting results concerning the role of iron in atherogenesis from several epidemiological and animal studies. We argue that atherosclerosis is not observed in HH because iron homeostasis in the arterial wall, the actual location of atherosclerosis, is not significantly affected, and support a causal link between iron in the arterial wall and atherosclerosis.
Collapse
Affiliation(s)
- Qian Guo
- Institute of Translational & Precision Medicine, Nantong University, Nantong, JS 226001, China; School of Medicine, Shanghai University, Shanghai 200444, China
| | - Christopher Qian
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Zhong-Ming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, JS 226001, China; Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
13
|
Graham FJ, Pellicori P, Kalra PR, Ford I, Bruzzese D, Cleland JG. Intravenous iron in patients with heart failure and iron deficiency: an updated meta-analysis. Eur J Heart Fail 2023; 25:528-537. [PMID: 36823953 PMCID: PMC10946839 DOI: 10.1002/ejhf.2810] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/04/2023] [Accepted: 02/22/2023] [Indexed: 02/25/2023] Open
Abstract
AIMS For patients with heart failure (HF) and iron deficiency (ID), randomized trials suggest that intravenous (IV) iron reduces hospitalizations for heart failure (HHF), but uncertainty exists about the effects in subgroups and the impact on mortality. We conducted a meta-analysis of randomized trials investigating the effect of IV iron on clinical outcomes in patients with HF. METHODS AND RESULTS We identified randomized trials published between 1 January 2000 and 5 November 2022 investigating the effect of IV iron versus standard care/placebo in patients with HF and ID in any clinical setting, regardless of HF phenotype. Trials of oral iron or not in English were not included. The main outcomes of interest were a composite of HHF and cardiovascular death (CVD), on HHF alone and on cardiovascular and all-cause mortality. Ten trials were identified with 3373 participants, of whom 1759 were assigned to IV iron. IV iron reduced the composite of recurrent HHF and CVD (rate ratio 0.75, 95% confidence interval [CI] 0.61-0.93; p < 0.01) and first HHF or CVD (odds ratio [OR] 0.72, 95% CI 0.53-0.99; p = 0.04). Effects on cardiovascular (OR 0.86, 95% CI 0.70-1.05; p = 0.14) and all-cause mortality (OR 0.93, 95% CI 0.78-1.12; p = 0.47) were inconclusive. Results were similar in analyses confined to the first year of follow-up, which was less disrupted by the COVID-19 pandemic. Subgroup analyses found little evidence of heterogeneity for the effect on the primary endpoint, although patients with transferrin saturation <20% (OR 0.67, 95% CI 0.49-0.92) may have benefited more than those with values ≥20% (OR 0.99, 95% CI 0.74-1.30) (heterogeneity p = 0.07). CONCLUSION In patients with HF and ID, this meta-analysis suggests that IV iron reduces the risk of HHF but whether this is associated with a reduction in cardiovascular or all-cause mortality remains inconclusive.
Collapse
Affiliation(s)
- Fraser J. Graham
- Robertson Centre for BiostatisticsUniversity of GlasgowGlasgowUK
| | | | - Paul R. Kalra
- Department of CardiologyPortsmouth Hospitals University NHS TrustPortsmouthUK
- College of Medical, Veterinary & Life SciencesUniversity of GlasgowGlasgowUK
- Faculty of Science and HealthUniversity of PortsmouthPortsmouthUK
| | - Ian Ford
- Robertson Centre for BiostatisticsUniversity of GlasgowGlasgowUK
| | - Dario Bruzzese
- Department of Public HealthUniversity of Naples Federico IINaplesItaly
| | - John G.F. Cleland
- School of Cardiovascular and Metabolic HealthUniversity of GlasgowGlasgowUK
| |
Collapse
|
14
|
Fratta Pasini AM, Stranieri C, Busti F, Di Leo EG, Girelli D, Cominacini L. New Insights into the Role of Ferroptosis in Cardiovascular Diseases. Cells 2023; 12:cells12060867. [PMID: 36980208 PMCID: PMC10047059 DOI: 10.3390/cells12060867] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the principal cause of disease burden and death worldwide. Ferroptosis is a new form of regulated cell death mainly characterized by altered iron metabolism, increased polyunsaturated fatty acid peroxidation by reactive oxygen species, depletion of glutathione and inactivation of glutathione peroxidase 4. Recently, a series of studies have indicated that ferroptosis is involved in the death of cardiac and vascular cells and has a key impact on the mechanisms leading to CVDs such as ischemic heart disease, ischemia/reperfusion injury, cardiomyopathies, and heart failure. In this article, we reviewed the molecular mechanism of ferroptosis and the current understanding of the pathophysiological role of ferroptosis in ischemic heart disease and in some cardiomyopathies. Moreover, the comprehension of the machinery governing ferroptosis in vascular cells and cardiomyocytes may provide new insights into preventive and therapeutic strategies in CVDs.
Collapse
|
15
|
Wang L, Cai J, Qiao T, Li K. Ironing out macrophages in atherosclerosis. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1-10. [PMID: 36647723 PMCID: PMC10157607 DOI: 10.3724/abbs.2022196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
<p indent="0mm">The most common cause of death worldwide is atherosclerosis and related cardiovascular disorders. Macrophages are important players in the pathogenesis of atherosclerosis and perform critical functions in iron homeostasis due to recycling iron by phagocytosis of senescent red blood cells and regulating iron availability in the tissue microenvironment. With the growth of research on the "iron hypothesis" of atherosclerosis, macrophage iron has gradually become a hotspot in the refined iron hypothesis. Macrophages with the M1, M2, M(Hb), Mox, and other phenotypes have been defined with different iron-handling capabilities related to the immune function and immunometabolism of macrophages, which influence the progression of atherosclerosis. In this review, we focus on macrophage iron and its effects on the development of atherosclerosis. We also cover the contradictory discoveries and propose a possible explanation. Finally, pharmaceutical modulation of macrophage iron is discussed as a promising target for atherosclerosis therapy.</p>.
Collapse
Affiliation(s)
- Lei Wang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jing Cai
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Tong Qiao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Kuanyu Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| |
Collapse
|
16
|
Teng T, Kong CY, Huang R, Ma ZG, Hu C, Zhang X, Hu M, Tang QZ. Mapping current research and identifying hotspots of ferroptosis in cardiovascular diseases. Front Cardiovasc Med 2022; 9:1046377. [PMID: 36407433 PMCID: PMC9672080 DOI: 10.3389/fcvm.2022.1046377] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/19/2022] [Indexed: 07/21/2023] Open
Abstract
OBJECTIVE Ferroptosis is a unique cell death depended on iron metabolism disorder which is different from previous apoptosis-regulated cell death. Early studies have proposed that ferroptosis is closely associated with multiple cardiovascular diseases (CVDs). However, the relationship of ferroptosis and CVDs has not been summarized by using bibliometric analysis. We intended to illustrate the development of ferroptosis in CVDs over the past years and provide relevant valuable information. MATERIALS AND METHODS The authoritative database of Web of Science Core Collection was collected for retrieving ferroptosis studies in CVDs. In this work, statistical and visualization analysis were conducted using VOSviewer and Citespace. RESULTS A total of 263 studies were included in the final study. From the perspective of the overall literature, the study maintains an increased trend year by year and most manuscripts belonged to original article. China was the most productive country with the utmost scientific research output, as well as the institutions and authors, followed by Germany and the United States of America (USA). Jun Peng from China contributes to the most publications. Collaborative efforts between institutes and authors were limited and there was little widespread cooperation. In addition, burst keywords analysis discovered that ischemia-reperfusion (I/R) injury, heart failure (HF), and atherosclerosis were the top three research directions of ferroptosis in CVDs. The burst investigation and timeline views also indicated that endothelial injury and gut microbiota may also serve as new research topics in the future. CONCLUSION This study provided comprehensive and specific information about the most influential articles on ferroptosis in CVDs. The relationship between ferroptosis and CVDs had attracted the scholar's concerns especially in China. Cooperations and communications between countries and institutions should be emphasized and future directions can be concentrated on endothelial disorder and gut microbiota.
Collapse
Affiliation(s)
- Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Chun-Yan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Rong Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Can Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Min Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| |
Collapse
|
17
|
Jamialahmadi T, Abbasifard M, Reiner Ž, Rizzo M, Eid AH, Sahebkar A. The Effects of Statin Treatment on Serum Ferritin Levels: A Systematic Review and Meta-Analysis. J Clin Med 2022; 11:5251. [PMID: 36079181 PMCID: PMC9457426 DOI: 10.3390/jcm11175251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Statins are the most widely used drugs for decreasing elevated serum LDL-cholesterol (LDL-C) and thus for the prevention of atherosclerotic cardiovascular disease (ASCVD), but they have also some pleiotropic effects, including anti-inflammatory properties. Atherosclerosis is a low-grade inflammatory disease, and elevated ferritin is considered to be one of the markers of inflammation. Since the results of studies on the effects of statins on serum ferritin levels are conflicting, this meta-analysis was performed. METHODS A literature search was performed using major electronic databases (MEDLINE/PubMed, Scopus, Embase, and ISI Web of Science) from inception up to 5 March 2022 to find studies evaluating the effect of different statins on serum ferritin levels. The effect size was determined using weighted mean differences (WMDs) and the corresponding 95% confidence intervals (CIs). RESULTS The meta-analysis of nine studies (1611 patients) analyzing the effects of statins on serum ferritin levels that were included showed a significant decrease in circulating ferritin levels caused by statins. The results did not suggest any significant association between the changes in concentrations of serum ferritin and the duration of treatment with statins. CONCLUSIONS Statin therapy decreases the circulating concentrations of ferritin, which might be beneficial for the prevention and/or progression of ASCVD. This effect might be explained by the anti-inflammatory effects and maybe some other pleiotropic effects of statins and not by their lipid-lowering effects.
Collapse
Affiliation(s)
- Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Mitra Abbasifard
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, 90133 Palermo, Italy
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
18
|
Li M, Wang ZW, Fang LJ, Cheng SQ, Wang X, Liu NF. Programmed cell death in atherosclerosis and vascular calcification. Cell Death Dis 2022; 13:467. [PMID: 35585052 PMCID: PMC9117271 DOI: 10.1038/s41419-022-04923-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/30/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022]
Abstract
The concept of cell death has been expanded beyond apoptosis and necrosis to additional forms, including necroptosis, pyroptosis, autophagy, and ferroptosis. These cell death modalities play a critical role in all aspects of life, which are noteworthy for their diverse roles in diseases. Atherosclerosis (AS) and vascular calcification (VC) are major causes for the high morbidity and mortality of cardiovascular disease. Despite considerable advances in understanding the signaling pathways associated with AS and VC, the exact molecular basis remains obscure. In the article, we review the molecular mechanisms that mediate cell death and its implications for AS and VC. A better understanding of the mechanisms underlying cell death in AS and VC may drive the development of promising therapeutic strategies.
Collapse
Affiliation(s)
- Min Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Zhen-Wei Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Li-Juan Fang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Shou-Quan Cheng
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Xin Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China.
| |
Collapse
|
19
|
Dos Santos L, Bertoli SR, Ávila RA, Marques VB. Iron overload, oxidative stress and vascular dysfunction: Evidences from clinical studies and animal models. Biochim Biophys Acta Gen Subj 2022; 1866:130172. [PMID: 35597504 DOI: 10.1016/j.bbagen.2022.130172] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 03/07/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022]
Abstract
Although iron is a metal involved in many in vital processes due to its redox capacity, body iron overloads lead to tissue damage, including the cardiovascular system. While cardiomyopathy was the focus since the 1960s, the impact on the vasculature was comparatively neglected for about 40 years, when clinical studies correlating iron overload, oxidative stress, endothelial dysfunction, arterial stiffness and atherosclerosis reinforced an "iron hypothesis". Due to controversial results from some epidemiological studies investigating atherosclerotic events and iron levels, well-controlled trials and animal studies provided essential data about the influence of iron, per se, on the vasculature. As a result, the pathophysiology of vascular dysfunction in iron overload have been revisited. This review summarizes the knowledge obtained from epidemiological studies, animal models and "in vitro" cellular systems in recent decades, highlighting a more harmful than innocent role of iron excess for the vascular homeostasis, which supports our proposal to hereafter denominate "iron overload vasculopathy". Additionally, evidence-based therapeutic targets are pointed out to be tested in pre-clinical research that may be useful in cardiovascular protection for patients with iron overload syndromes.
Collapse
Affiliation(s)
- Leonardo Dos Santos
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, ES, Brazil.
| | - Sabrina Rodrigues Bertoli
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, ES, Brazil; Faculdade Novo Milenio, Vila Velha, ES, Brazil
| | - Renata Andrade Ávila
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, ES, Brazil; Faculdades Integradas São Pedro (FAESA), Vitória, ES, Brazil
| | | |
Collapse
|
20
|
Fernández-García V, González-Ramos S, Avendaño-Ortiz J, Martín-Sanz P, Delgado C, Castrillo A, Boscá L. NOD1 splenic activation confers ferroptosis protection and reduces macrophage recruitment under pro-atherogenic conditions. Biomed Pharmacother 2022; 148:112769. [PMID: 35247718 DOI: 10.1016/j.biopha.2022.112769] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/15/2022] [Accepted: 02/27/2022] [Indexed: 02/07/2023] Open
Abstract
The bioavailability and regulation of iron is essential for central biological functions in mammals. The role of this element in ferroptosis and the dysregulation of its metabolism contribute to diseases, ranging from anemia to infections, alterations in the immune system, inflammation and atherosclerosis. In this sense, monocytes and macrophages modulate iron metabolism and splenic function, while at the same time they can worsen the atherosclerotic process in pathological conditions. Since the nucleotide-binding oligomerization domain 1 (NOD1) has been linked to numerous disorders, including inflammatory and cardiovascular diseases, we investigated its role in iron homeostasis. The iron content was measured in various tissues of Apoe-/- and Apoe-/-Nod1-/- mice fed a high-fat diet (HFD) for 4 weeks, under normal or reduced splenic function after ligation of the splenic artery. In the absence of NOD1 the iron levels decreased in spleen, heart and liver regardless the splenic function. This iron decrease was accompanied by an increase in the recruitment of F4/80+-macrophages in the spleen through a CXCR2-dependent signaling, as deduced by the reduced recruitment after administration of a CXCR2 inhibitor. CXCR2 mediates monocyte/macrophage chemotaxis to areas of inflammation and accumulation of leukocytes in the atherosclerotic plaque. Moreover, in the absence of NOD1, inhibition of CXCR2 enhanced atheroma progression. NOD1 activation increased the levels of GPX4 and other iron and ferroptosis regulatory proteins in macrophages. Our findings highlight the preeminent role of NOD1 in iron homeostasis and ferroptosis. These results suggest promising avenues of investigation for the diagnosis and treatment of iron-related diseases directed by NOD1.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid 28029, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, Madrid 28029, Spain.
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid 28029, Spain
| | - José Avendaño-Ortiz
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz, IdiPAZ. Pedro Rico, 6, Madrid 28029, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid 28029, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid 28029, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid 28029, Spain; Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid 28029, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, Madrid 28029, Spain.
| |
Collapse
|
21
|
Guo Y, Lu C, Hu K, Cai C, Wang W. Ferroptosis in Cardiovascular Diseases: Current Status, Challenges, and Future Perspectives. Biomolecules 2022; 12:biom12030390. [PMID: 35327582 PMCID: PMC8945958 DOI: 10.3390/biom12030390] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular diseases (CVDs) are still a major cause of global mortality and disability, seriously affecting people’s lives. Due to the severity and complexity of these diseases, it is important to find new regulatory mechanisms to treat CVDs. Ferroptosis is a new kind of regulatory cell death currently being investigated. Increasing evidence showed that ferroptosis plays an important role in CVDs, such as in ischemia/reperfusion injury, heart failure, cardiomyopathy, and atherosclerosis. Protecting against CVDs by targeting ferroptosis is a promising approach; therefore, in this review, we summarized the latest regulatory mechanism of ferroptosis and the current studies related to each CVD, followed by critical perspectives on the ferroptotic treatment of CVDs and the future direction of this intriguing biology.
Collapse
Affiliation(s)
- Yi Guo
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
| | - Chanjun Lu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
| | - Ke Hu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
| | - Weici Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
- Correspondence: ; Tel.: +86-180-7170-5166
| |
Collapse
|
22
|
Zhang Z, Liu C, Liu M, Zhou C, Li Q, He P, Zhang Y, Li H, Qin X. Dietary Iron Intake and New-Onset Hypertension: A Nationwide Cohort Study from China. J Nutr Health Aging 2022; 26:1016-1024. [PMID: 36437770 DOI: 10.1007/s12603-022-1861-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The relationship of dietary iron intake with the risk of hypertension remains uncertain. We aimed to investigate the prospective association between dietary iron intake and new-onset hypertension among Chinese adults. DESIGN A nationwide cohort study. SETTING Using data from seven rounds of the China Health and Nutrition Survey (CHNS) from 1997 to 2015. PARTICIPANTS A total of 12,245 participants without hypertension at baseline were included in this study. EXPOSURES Dietary intake was measured by three consecutive 24-h dietary recalls combined with a household weighing inventory. MEASUREMENTS The study outcome was new-onset hypertension, defined as systolic blood pressure ≥140 mmHg and/or diastolic blood pressure ≥90 mmHg or diagnosed by physician or currently under antihypertensive treatment during the follow-up. RESULTS During a median follow-up of 6.1 years, 4,304 (35.1%) participants developed hypertension. Overall, there was a U-shaped association between dietary total iron intake and new-onset hypertension (P for nonlinearity <0.001), with the lowest risk observed at 18.2-<22.1 mg/day (quintile 2-3). Similarly, a U-shaped association between dietary nonheme iron intake and new-onset hypertension was found (P for nonlinearity <0.001), with the lowest risk at 17.4-<21.3 mg/day (quintile 2-3). However, the association between dietary heme iron intake and new-onset hypertension followed a L-shape (P for nonlinearity <0.001), and a significantly lower risk of new-onset hypertension was found in participants with quintile 2-5 of dietary heme iron intake (adjusted HR, 0.75; 95% CI: 0.68, 0.82), compared with those in quintile 1 (<0.25 mg/day). CONCLUSIONS The association between dietary iron and new-onset hypertension was nonlinear in Chinese adults, following a U-shape for total or nonheme iron intake, and a L-shape for heme iron intake.
Collapse
Affiliation(s)
- Z Zhang
- Xianhui Qin, M.D., Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; ; Phone: 86-20-61641591; Fax: 86-20-87281713
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Trichoderma and Its Products From Laboratory to Patient Bedside in Medical Science: An Emerging Aspect. Fungal Biol 2022. [DOI: 10.1007/978-3-030-91650-3_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
24
|
Li JY, Liu SQ, Yao RQ, Tian YP, Yao YM. A Novel Insight Into the Fate of Cardiomyocytes in Ischemia-Reperfusion Injury: From Iron Metabolism to Ferroptosis. Front Cell Dev Biol 2021; 9:799499. [PMID: 34926476 PMCID: PMC8675329 DOI: 10.3389/fcell.2021.799499] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Ischemia-reperfusion injury (IRI), critically involved in the pathology of reperfusion therapy for myocardial infarction, is closely related to oxidative stress the inflammatory response, and disturbances in energy metabolism. Emerging evidence shows that metabolic imbalances of iron participate in the pathophysiological process of cardiomyocyte IRI [also termed as myocardial ischemia-reperfusion injury (MIRI)]. Iron is an essential mineral required for vital physiological functions, including cellular respiration, lipid and oxygen metabolism, and protein synthesis. Nevertheless, cardiomyocyte homeostasis and viability are inclined to be jeopardized by iron-induced toxicity under pathological conditions, which is defined as ferroptosis. Upon the occurrence of IRI, excessive iron is transported into cells that drive cardiomyocytes more vulnerable to ferroptosis by the accumulation of reactive oxygen species (ROS) through Fenton reaction and Haber–Weiss reaction. The increased ROS production in ferroptosis correspondingly leads cardiomyocytes to become more sensitive to oxidative stress under the exposure of excess iron. Therefore, ferroptosis might play an important role in the pathogenic progression of MIRI, and precisely targeting ferroptosis mechanisms may be a promising therapeutic option to revert myocardial remodeling. Notably, targeting inhibitors are expected to prevent MIRI deterioration by suppressing cardiomyocyte ferroptosis. Here, we review the pathophysiological alterations from iron homeostasis to ferroptosis together with potential pathways regarding ferroptosis secondary to cardiovascular IRI. We also provide a comprehensive analysis of ferroptosis inhibitors and initiators, as well as regulatory genes involved in the setting of MIRI.
Collapse
Affiliation(s)
- Jing-yan Li
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuang-qing Liu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Ren-qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Ying-ping Tian
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Ying-ping Tian, ; Yong-ming Yao,
| | - Yong-ming Yao
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- *Correspondence: Ying-ping Tian, ; Yong-ming Yao,
| |
Collapse
|
25
|
Luo MY, Su JH, Gong SX, Liang N, Huang WQ, Chen W, Wang AP, Tian Y. Ferroptosis: New Dawn for Overcoming the Cardio-Cerebrovascular Diseases. Front Cell Dev Biol 2021; 9:733908. [PMID: 34858973 PMCID: PMC8632439 DOI: 10.3389/fcell.2021.733908] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/25/2021] [Indexed: 12/21/2022] Open
Abstract
The dynamic balance of cardiomyocytes and neurons is essential to maintain the normal physiological functions of heart and brain. If excessive cells die in tissues, serious Cardio-Cerebrovascular Diseases would occur, namely, hypertension, myocardial infarction, and ischemic stroke. The regulation of cell death plays a role in promoting or alleviating Cardio-Cerebrovascular Diseases. Ferroptosis is an iron-dependent new type of cell death that has been proved to occur in a variety of diseases. In our review, we focus on the critical role of ferroptosis and its regulatory mechanisms involved in Cardio-Cerebrovascular Diseases, and discuss the important function of ferroptosis-related inhibitors in order to propose potential implications for the prevention and treatment of Cardio-Cerebrovascular Diseases.
Collapse
Affiliation(s)
- Meng-Yi Luo
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Jian-Hui Su
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, University of South China, Hengyang, China
| | - Na Liang
- Department of Anesthesiology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Wen-Qian Huang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Wei Chen
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| |
Collapse
|
26
|
Alonso-Piñeiro JA, Gonzalez-Rovira A, Sánchez-Gomar I, Moreno JA, Durán-Ruiz MC. Nrf2 and Heme Oxygenase-1 Involvement in Atherosclerosis Related Oxidative Stress. Antioxidants (Basel) 2021; 10:1463. [PMID: 34573095 PMCID: PMC8466960 DOI: 10.3390/antiox10091463] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis remains the underlying process responsible for cardiovascular diseases and the high mortality rates associated. This chronic inflammatory disease progresses with the formation of occlusive atherosclerotic plaques over the inner walls of vascular vessels, with oxidative stress being an important element of this pathology. Oxidation of low-density lipoproteins (ox-LDL) induces endothelial dysfunction, foam cell activation, and inflammatory response, resulting in the formation of fatty streaks in the atherosclerotic wall. With this in mind, different approaches aim to reduce oxidative damage as a strategy to tackle the progression of atherosclerosis. Special attention has been paid in recent years to the transcription factor Nrf2 and its downstream-regulated protein heme oxygenase-1 (HO-1), both known to provide protection against atherosclerotic injury. In the current review, we summarize the involvement of oxidative stress in atherosclerosis, focusing on the role that these antioxidant molecules exert, as well as the potential therapeutic strategies applied to enhance their antioxidant and antiatherogenic properties.
Collapse
Affiliation(s)
- Jose Angel Alonso-Piñeiro
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, 11519 Puerto Real, Spain; (J.A.A.-P.); (A.G.-R.); (I.S.-G.)
- Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), 11001 Cádiz, Spain
| | - Almudena Gonzalez-Rovira
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, 11519 Puerto Real, Spain; (J.A.A.-P.); (A.G.-R.); (I.S.-G.)
- Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), 11001 Cádiz, Spain
| | - Ismael Sánchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, 11519 Puerto Real, Spain; (J.A.A.-P.); (A.G.-R.); (I.S.-G.)
- Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), 11001 Cádiz, Spain
| | - Juan Antonio Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), UGC Nephrology, Hospital Universitario Reina Sofia, 14004 Cordoba, Spain;
- Department of Cell Biology, Physiology, and Immunology, Agrifood Campus of International Excellence (ceiA3), University of Cordoba, 14014 Cordoba, Spain
| | - Ma Carmen Durán-Ruiz
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, 11519 Puerto Real, Spain; (J.A.A.-P.); (A.G.-R.); (I.S.-G.)
- Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), 11001 Cádiz, Spain
| |
Collapse
|
27
|
Zingg JM, Vlad A, Ricciarelli R. Oxidized LDLs as Signaling Molecules. Antioxidants (Basel) 2021; 10:antiox10081184. [PMID: 34439432 PMCID: PMC8389018 DOI: 10.3390/antiox10081184] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Levels of oxidized low-density lipoproteins (oxLDLs) are usually low in vivo but can increase whenever the balance between formation and scavenging of free radicals is impaired. Under normal conditions, uptake and degradation represent the physiological cellular response to oxLDL exposure. The uptake of oxLDLs is mediated by cell surface scavenger receptors that may also act as signaling molecules. Under conditions of atherosclerosis, monocytes/macrophages and vascular smooth muscle cells highly exposed to oxLDLs tend to convert to foam cells due to the intracellular accumulation of lipids. Moreover, the atherogenic process is accelerated by the increased expression of the scavenger receptors CD36, SR-BI, LOX-1, and SRA in response to high levels of oxLDL and oxidized lipids. In some respects, the effects of oxLDLs, involving cell proliferation, inflammation, apoptosis, adhesion, migration, senescence, and gene expression, can be seen as an adaptive response to the rise of free radicals in the vascular system. Unlike highly reactive radicals, circulating oxLDLs may signal to cells at more distant sites and possibly trigger a systemic antioxidant defense, thus elevating the role of oxLDLs to that of signaling molecules with physiological relevance.
Collapse
Affiliation(s)
- Jean-Marc Zingg
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Correspondence: (J.-M.Z.); (R.R.); Tel.: +1-(305)-2433531 (J.-M.Z.); +39-010-3538831 (R.R.)
| | - Adelina Vlad
- Physiology Department, “Carol Davila” UMPh, 020021 Bucharest, Romania;
| | - Roberta Ricciarelli
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence: (J.-M.Z.); (R.R.); Tel.: +1-(305)-2433531 (J.-M.Z.); +39-010-3538831 (R.R.)
| |
Collapse
|
28
|
The rs508487, rs236911, and rs236918 Genetic Variants of the Proprotein Convertase Subtilisin-Kexin Type 7 ( PCSK7) Gene Are Associated with Acute Coronary Syndrome and with Plasma Concentrations of HDL-Cholesterol and Triglycerides. Cells 2021; 10:cells10061444. [PMID: 34207761 PMCID: PMC8227151 DOI: 10.3390/cells10061444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/01/2021] [Accepted: 06/05/2021] [Indexed: 01/18/2023] Open
Abstract
Dyslipidemia has a substantial role in the development of acute coronary syndrome (ACS). Previous reports, including genome-wide associations studies (GWAS), have shown that some genetic variants of the proprotein convertase subtilisin–kexin type 7 (PCSK7) gene are associated with plasma lipid levels. In the present study, we evaluated whether PCSK7 gene polymorphisms are significantly associated with the plasma lipid profile and ACS. Three PCSK7 gene polymorphisms (rs508487 T/C, rs236911 C/A, and rs236918 C/G) were determined using TaqMan genotyping assays in a group of 603 ACS patients and 622 healthy controls. The plasma lipid profile was determined in the study groups by enzymatic/colorimetric assays. Under the recessive model, the rs236918 C allele was associated with a high risk of ACS (OR = 2.11, pC = 0.039). In the same way, under the recessive and additive models, the rs236911 C allele was associated with a high risk of ACS (OR = 1.95, pC = 0.037, and OR = 1.28, pC = 0.037, respectively). In addition, under the co-dominant model, the rs508487 T allele was associated with a higher risk of ACS (OR = 1.78, pC = 0.010). The CCC and TCC haplotypes were associated with a high risk of ACS (OR = 1.21, pC = 0.047, and OR = 1.80, pC = 0.001, respectively). The rs236911 CC and rs236918 CC genotypes were associated with lower high-density lipoproteins-cholesterol (HDL-C) plasma concentrations, whereas the rs236911 CC genotype was associated with a higher concentration of triglycerides, as demonstrated in the control individuals who were not receiving antidyslipidemic drugs. Our data suggest that the PCSK7 rs508487 T/C, rs236911 C/A, and rs236918 C/G polymorphisms are associated with the risk of developing ACS, and with plasma concentrations of HDL-C and triglycerides.
Collapse
|
29
|
Turpin C, Catan A, Meilhac O, Bourdon E, Canonne-Hergaux F, Rondeau P. Erythrocytes: Central Actors in Multiple Scenes of Atherosclerosis. Int J Mol Sci 2021; 22:ijms22115843. [PMID: 34072544 PMCID: PMC8198892 DOI: 10.3390/ijms22115843] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022] Open
Abstract
The development and progression of atherosclerosis (ATH) involves lipid accumulation, oxidative stress and both vascular and blood cell dysfunction. Erythrocytes, the main circulating cells in the body, exert determinant roles in the gas transport between tissues. Erythrocytes have long been considered as simple bystanders in cardiovascular diseases, including ATH. This review highlights recent knowledge concerning the role of erythrocytes being more than just passive gas carriers, as potent contributors to atherosclerotic plaque progression. Erythrocyte physiology and ATH pathology is first described. Then, a specific chapter delineates the numerous links between erythrocytes and atherogenesis. In particular, we discuss the impact of extravasated erythrocytes in plaque iron homeostasis with potential pathological consequences. Hyperglycaemia is recognised as a significant aggravating contributor to the development of ATH. Then, a special focus is made on glycoxidative modifications of erythrocytes and their role in ATH. This chapter includes recent data proposing glycoxidised erythrocytes as putative contributors to enhanced atherothrombosis in diabetic patients.
Collapse
Affiliation(s)
- Chloé Turpin
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Aurélie Catan
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Olivier Meilhac
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Centre Hospitalier Universitaire de La Réunion, 97400 Saint Denis, France
| | - Emmanuel Bourdon
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | | | - Philippe Rondeau
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Correspondence: ; Tel.: +262(0)-2-62-93-88-43; Fax: +262-(0)-2-62-93-88-01
| |
Collapse
|
30
|
Xia Y, Li Y, Wu X, Zhang Q, Chen S, Ma X, Yu M. Ironing Out the Details: How Iron Orchestrates Macrophage Polarization. Front Immunol 2021; 12:669566. [PMID: 34054839 PMCID: PMC8149954 DOI: 10.3389/fimmu.2021.669566] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Iron fine-tunes innate immune responses, including macrophage inflammation. In this review, we summarize the current understanding about the iron in dictating macrophage polarization. Mechanistically, iron orchestrates macrophage polarization through several aspects, including cellular signaling, cellular metabolism, and epigenetic regulation. Therefore, iron modulates the development and progression of multiple macrophage-associated diseases, such as cancer, atherosclerosis, and liver diseases. Collectively, this review highlights the crucial role of iron for macrophage polarization, and indicates the potential application of iron supplementation as an adjuvant therapy in different inflammatory disorders relative to the balance of macrophage polarization.
Collapse
Affiliation(s)
- Yaoyao Xia
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yikun Li
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaoyan Wu
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qingzhuo Zhang
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siyuan Chen
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xianyong Ma
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Miao Yu
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
31
|
Xu Y, Wang Y, Hu H, Li J, Tian T. Relationship between serum hepcidin levels and cardiovascular disease in patients with maintenance hemodialysis. Physiol Int 2020; 107:491-500. [PMID: 33355540 DOI: 10.1556/2060.2020.00040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/06/2020] [Indexed: 11/19/2022]
Abstract
Background To investigate the serum level of hepcidin and its relationship with cardiovascular disease (CVD) in maintenance hemodialysis (MHD) patients. Methods Blood was obtained from 75 MHD patients before undergoing hemodialysis and 20 healthy controls. Serum hepcidin, advanced oxidation protein products (AOPP) and interleukin (IL)-6 were measured by enzyme-linked immunosorbant assay (ELISA). Spearman correlation, and binary logistic regression linear regression analyses were used to assess the relationship between serum hepcidin and other parameters. Results The serum level of hepcidin, AOPP and IL-6 was significantly up-regulated in MHD patients compared with the control (P < 0.05). Furthermore, serum hepcidin levels in patients with CVD were higher than those in patients without CVD (P < 0.05). In all MHD patients, serum hepcidin level was correlated positively with erythropoietin (EPO) dose per week (ρ = 0.251, P = 0.030), EPO resistance index (ρ = 0.268, P = 0.020), ferritin (ρ = 0.814, P < 0.001), transferin saturation (TSAT, ρ = 0.263, P = 0.023), AOPP (ρ = 0.280, P = 0.049), high sensitive C reactive protein (ρ = 0.151, P = 0.006), IL-6 (ρ = 0.340, P = 0.003) and left ventricular mass index (LVMI, ρ = 0.290, P = 0.033). Moreover, it was negatively correlated with serum pre-albumin (ρ = -0.266, P = 0.021), total iron-binding capacity (TIBC, ρ = -0.458, P < 0.001), unsaturated iron-binding capacity (UIBC, ρ = -0.473, P < 0.001) and transferrin (ρ = -0.487, P < 0.001). Linear regression analysis showed that ferritin (β = 0.708, P < 0.001), TIBC (β = -0.246, P = 0.032) and IL-6 (β = 0.209, P = 0.041) were independently associated with hepcidin. Results of binary logistic regression analysis suggested that higher serum hepcidin level (>249.2 ng/mL) was positively and independently related to CVD (OR = 1.32, 95% CI [1.20-9.56], P = 0.043). Conclusions Serum hepcidin level is associated with CVD in MHD patients, indicating that hepcidin may be a novel biomarker and therapeutic target for CVD.
Collapse
Affiliation(s)
- Y Xu
- 1Hemodialysis Center, Shanghai Yangsi Hospital, Pudong New Area, Shanghai, China
| | - Y Wang
- 2Hemodialysis Center, Yantai Affiliated Hospital of Binzhou Medical University, Yantai City, Shandong Province, China
| | - H Hu
- 1Hemodialysis Center, Shanghai Yangsi Hospital, Pudong New Area, Shanghai, China
| | - J Li
- 1Hemodialysis Center, Shanghai Yangsi Hospital, Pudong New Area, Shanghai, China
| | - T Tian
- 1Hemodialysis Center, Shanghai Yangsi Hospital, Pudong New Area, Shanghai, China
| |
Collapse
|
32
|
Cai J, Zhang M, Liu Y, Li H, Shang L, Xu T, Chen Z, Wang F, Qiao T, Li K. Iron accumulation in macrophages promotes the formation of foam cells and development of atherosclerosis. Cell Biosci 2020; 10:137. [PMID: 33292517 PMCID: PMC7691057 DOI: 10.1186/s13578-020-00500-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Macrophages that accumulate in atherosclerotic plaques contribute to progression of the lesions to more advanced and complex plaques. Although iron deposition was found in human atherosclerotic plaques, clinical and pre-clinical studies showed controversial results. Several epidemiological studies did not show the positive correlation between a systemic iron status and an incidence of cardiovascular diseases, suggesting that the iron involvement occurs locally, rather than systemically. RESULTS To determine the direct in vivo effect of iron accumulation in macrophages on the progression of atherosclerosis, we generated Apoe-/- mice with a macrophage-specific ferroportin (Fpn1) deficiency (Apoe-/-Fpn1LysM/LysM). Fpn1 deficiency in macrophages dramatically accelerated the progression of atherosclerosis in mice. Pathophysiological evidence showed elevated levels of reactive oxygen species, aggravated systemic inflammation, and altered plaque-lipid composition. Moreover, Fpn1 deficiency in macrophages significantly inhibited the expression of ABC transporters (ABCA1 and ABCG1) by decreasing the expression of the transcription factor LXRα, which reduced cholesterol efflux and therefore promoted foam cell formation and enhanced plaque formation. Iron chelation relieved the symptoms moderately in vivo, but drastically ex vivo. CONCLUSIONS Macrophage iron content in plaques is a critical factor in progression of atherosclerosis. The interaction of iron and lipid metabolism takes place in macrophage-rich atherosclerotic plaques. And we also suggest that altering intracellular iron levels in macrophages by systemic iron chelation or dietary iron restriction may be a potential supplementary strategy to limit or even regress the progression of atherosclerosis.
Collapse
Affiliation(s)
- Jing Cai
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Meng Zhang
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Yutong Liu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, People's Republic of China
| | - Huihui Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, People's Republic of China
| | - Longcheng Shang
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Tianze Xu
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Zhipeng Chen
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Fudi Wang
- Department of Nutrition, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tong Qiao
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China.
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, People's Republic of China.
| |
Collapse
|
33
|
Bai T, Li M, Liu Y, Qiao Z, Wang Z. Inhibition of ferroptosis alleviates atherosclerosis through attenuating lipid peroxidation and endothelial dysfunction in mouse aortic endothelial cell. Free Radic Biol Med 2020; 160:92-102. [PMID: 32768568 DOI: 10.1016/j.freeradbiomed.2020.07.026] [Citation(s) in RCA: 332] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/24/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022]
Abstract
Atherosclerosis (AS) is the fundamental pathological state of many serious vascular diseases, characterized by disorders of lipid metabolism. Ferroptosis is a type of regulated cell death that is mainly mediated by iron-dependent lipid peroxidation. In this study, whether ferroptosis has occurred in AS and the potential effects of ferroptosis on AS were investigated. Ferroptosis inhibitor ferrostatin-1 (Fer-1) was administered to high-fat diet (HFD)-induced AS in ApoE-/- mice. The results showed that Fer-1 could alleviate AS lesion in HFD-fed ApoE-/- mice. Additionally, Fer-1 partially inhibited the iron accumulation, lipid peroxidation and reversed the expressions of ferroptosis indicators SLC7A11 and glutathione peroxidase 4 (GPX4) in HFD-fed ApoE-/- mice. Next, we evaluated the effects of inhibition of ferroptosis on oxidized-low density lipoprotein (ox-LDL)-induced mouse aortic endothelial cells (MAECs). Results showed that Fer-1 increased cell viability and reduced cell death in ox-LDL-treated MAECs. Moreover, Fer-1 decreased iron content and lipid peroxidation and up-regulated the levels of SLC7A11 and GPX4. Additionally, Fer-1 down-regulated the expressions of adhesion molecules and up-regulated eNOS expression. Iron chelator deferoxamine was used to demonstrate ferroptosis could be partially inhibited by iron complexation in ox-LDL-treated MAECs. Our results indicated that ferroptosis might occur during the initiation and development of AS. More importantly, inhibition of ferroptosis could alleviate AS through attenuating lipid peroxidation and endothelial dysfunction in AECs. Our findings might contribute to a deeper understanding regarding the pathological process of AS and provide a therapeutic target for AS.
Collapse
Affiliation(s)
- Tao Bai
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Mingxing Li
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yuanfeng Liu
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zhentao Qiao
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zhiwei Wang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
34
|
The relationship between famine exposure during early life and carotid plaque in adulthood. Eur J Clin Nutr 2020; 75:546-554. [PMID: 32939040 DOI: 10.1038/s41430-020-00756-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 08/24/2020] [Accepted: 09/05/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Famine exposure is a potential risk factor for adverse cardiometabolic health. However, the relationship between famine exposure during early life and carotid plaque in adulthood remains unclear. Therefore, the aim was to investigate the relationship between famine exposure during early life and the risks for carotid plaque in adulthood. METHODS This was a cross-sectional study. Data were collected between 2017 and 2018 in Guangdong, China. Subjects who were born between 1 October 1952 and 30 September 1964, and had the carotid ultrasound measurement were enrolled. All included participants were divided into five groups: no exposure, fetal exposure, early-childhood exposure, mid-childhood exposure, and late-childhood exposure. Carotid plaque was assessed by carotid ultrasound examination. Multivariate logistic regression was used to estimate the odds ratio (OR) and confidence interval (CI) between famine exposure and carotid plaque. RESULTS There were 2652 subjects enrolled, 973 (36.7%) of them were males, and the mean age was 59.1 ± 3.6 years. The prevalence of carotid plaque in unexposed, fetal-exposed, early-childhood, mid-childhood, and late-childhood exposed groups were 40.2%, 40.8%, 55.3%, 56.8%, and 62.1%, respectively. When compared with the unexposed group, the fully adjusted ORs for carotid plaque from fetal-exposed, early-childhood, mid-childhood to late-childhood exposed were 1.023 (95% CI: 0.771, 1.357, P = 0.872), 1.755 (95% CI: 1.356, 2.275, P < 0.001), 1.780 (95% CI: 1.391, 2.280, P < 0.001), and 2.119 (95% CI: 1.643, 2.739, P < 0.001), respectively. Subgroup analyses showed that the famine effect on carotid plaque did not interact with body mass index, gender, smoking status, hypertension, and diabetes history (all P for interaction > 0.500). CONCLUSIONS Famine exposure during early life was significantly associated with an increased risk of carotid plaque in adulthood.
Collapse
|
35
|
Bosseboeuf E, Raimondi C. Signalling, Metabolic Pathways and Iron Homeostasis in Endothelial Cells in Health, Atherosclerosis and Alzheimer's Disease. Cells 2020; 9:cells9092055. [PMID: 32911833 PMCID: PMC7564205 DOI: 10.3390/cells9092055] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells drive the formation of new blood vessels in physiological and pathological contexts such as embryonic development, wound healing, cancer and ocular diseases. Once formed, all vessels of the vasculature system present an endothelial monolayer (the endothelium), lining the luminal wall of the vessels, that regulates gas and nutrient exchange between the circulating blood and tissues, contributing to maintaining tissue and vascular homeostasis. To perform their functions, endothelial cells integrate signalling pathways promoted by growth factors, cytokines, extracellular matrix components and signals from mechanosensory complexes sensing the blood flow. New evidence shows that endothelial cells rely on specific metabolic pathways for distinct cellular functions and that the integration of signalling and metabolic pathways regulates endothelial-dependent processes such as angiogenesis and vascular homeostasis. In this review, we provide an overview of endothelial functions and the recent advances in understanding the role of endothelial signalling and metabolism in physiological processes such as angiogenesis and vascular homeostasis and vascular diseases. Also, we focus on the signalling pathways promoted by the transmembrane protein Neuropilin-1 (NRP1) in endothelial cells, its recently discovered role in regulating mitochondrial function and iron homeostasis and the role of mitochondrial dysfunction and iron in atherosclerosis and neurodegenerative diseases.
Collapse
|
36
|
Rittase WB, Muir JM, Slaven JE, Bouten RM, Bylicky MA, Wilkins WL, Day RM. Deposition of Iron in the Bone Marrow of a Murine Model of Hematopoietic Acute Radiation Syndrome. Exp Hematol 2020; 84:54-66. [PMID: 32240658 DOI: 10.1016/j.exphem.2020.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/20/2020] [Accepted: 03/21/2020] [Indexed: 01/02/2023]
Abstract
Exposure to high-dose total body irradiation (TBI) can result in hematopoietic acute radiation syndrome (H-ARS), characterized by leukopenia, anemia, and coagulopathy. Death from H-ARS occurs from hematopoietic insufficiency and opportunistic infections. Following radiation exposure, red blood cells (RBCs) undergo hemolysis from radiation-induced hemoglobin denaturation, causing the release of iron. Free iron can have multiple detrimental biological effects, including suppression of hematopoiesis. We investigated the impact of radiation-induced iron release on the bone marrow following TBI and the potential impact of the ACE inhibitor captopril, which improves survival from H-ARS. C57BL/6J mice were exposed to 7.9 Gy, 60Co irradiation, 0.6 Gy/min (LD70-90/30). RBCs and reticulocytes were significantly reduced within 7 days of TBI, with the RBC nadir at 14-21 days. Iron accumulation in the bone marrow correlated with the time course of RBC hemolysis, with an ∼10-fold increase in bone marrow iron at 14-21 days post-irradiation, primarily within the cytoplasm of macrophages. Iron accumulation in the bone marrow was associated with increased expression of genes for iron binding and transport proteins, including transferrin, transferrin receptor 1, ferroportin, and integrin αMβ2. Expression of the gene encoding Nrf2, a transcription factor activated by oxidative stress, also increased at 21 days post-irradiation. Captopril did not alter iron accumulation in the bone marrow or expression of iron storage genes, but did suppress Nrf2 expression. Our study suggests that following TBI, iron is deposited in tissues not normally associated with iron storage, which may be a secondary mechanism of radiation-induced tissue injury.
Collapse
Affiliation(s)
- W Bradley Rittase
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Jeannie M Muir
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - John E Slaven
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Roxane M Bouten
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Michelle A Bylicky
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health Bethesda, MD
| | - W Louis Wilkins
- Department of Laboratory Animal Research, Uniformed Services University of Health Sciences, Bethesda, MD
| | - Regina M Day
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD.
| |
Collapse
|
37
|
Management of Iron Overload in Resource Poor Nations: A Systematic Review of Phlebotomy and Natural Chelators. J Toxicol 2020; 2020:4084538. [PMID: 32399029 PMCID: PMC7204175 DOI: 10.1155/2020/4084538] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/13/2019] [Accepted: 09/20/2019] [Indexed: 12/29/2022] Open
Abstract
Iron is an essential element and the most abundant trace metal in the body involved in oxygen transport and oxygen sensing, electron transfer, energy metabolism, and DNA synthesis. Excess labile and unchelated iron can catalyze the formation of tissue-damaging radicals and induce oxidative stress. English abstracts were identified in PubMed and Google Scholar using multiple and various search terms based on defined inclusion and exclusion criteria. Full-length articles were selected for systematic review, and secondary and tertiary references were developed. Although bloodletting or phlebotomy remains the gold standard in the management of iron overload, this systematic review is an updated account of the pitfalls of phlebotomy and classical synthetic chelators with scientific justification for the use of natural iron chelators of dietary origin in resource-poor nations.
Collapse
|
38
|
Xiao L, Luo G, Guo X, Jiang C, Zeng H, Zhou F, Li Y, Yu J, Yao P. Macrophage iron retention aggravates atherosclerosis: Evidence for the role of autocrine formation of hepcidin in plaque macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158531. [PMID: 31666189 DOI: 10.1016/j.bbalip.2019.158531] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 12/18/2022]
Abstract
Iron accumulation has been frequently found in atherosclerotic lesions, especially in macrophages/foam cells, but the exact mechanisms by which hepcidin induces iron retention in plaque macrophages and its roles in atherogenesis remain unknown. Double immunofluorescence staining showed colocalization of hepcidin-positive macrophages with ox-LDL, TLR4, p-p65 and ferritin light chain (ferritin-L) both in human and murine atherosclerotic lesions. RAW264.7 macrophages incubated with ox-LDL showed elevated expression of TLR4, p-p65, hepcidin, ferritin-L/H, CYP27A1, CD36, PPARγ, liver X receptor α (LXRα), and ATP binding cassette transporter A1/G1 (ABCA1/G1), as well as increased intracellular labile iron pool level and lipid accumulation. Ox-LDL-induced iron retention and lipid accumulation were aggravated by lipopolysaccharide but blocked by TAK-242, an antagonist of TLR4. Moreover, macrophage TLR4/NF-κB pathway activation and foaming triggered by ox-LDL was enhanced by ferric ammonium citrate or exogenous hepcidin but attenuated by hepcidin silencing or the use of iron chelator. Meanwhile, the addition of hepcidin stimulated CD36-mediated Dil-labeled-ox-LDL uptake and inhibited the LXRα-ABCA1/G1 pathway-dependent cholesterol efflux in macrophages, which was significantly reversed by 27-hydroxycholesterol but further exacerbated by cyclosporin A, a selective inhibitor of CYP27A1. Our study provided the evidence that iron trapped in atherosclerosis plaque macrophages contributes to cholesterol disequilibrium-initiated foam cell formation, which is provoked by the unique but largely unknown autocrine formation of hepcidin in plaque macrophages via activating the TLR4/NF-κB pathway when exposed to ox-LDL. Such findings, considering the intricate vicious cycle between macrophage hepcidin autocrine-triggered iron retention and cholesterol disequilibrium, may shed new light on the "iron hypothesis" of atherosclerosis.
Collapse
Affiliation(s)
- Lin Xiao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gang Luo
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoping Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chunjie Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongmei Zeng
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feng Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanyan Li
- Shenzhen Center for Chronic Disease Control, 2021 Buxin Road, Shenzhen 518020, China
| | - Jiasheng Yu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, HuaZhong University of Science and Technology, Wuhan 430030, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
39
|
Abstract
Iron is an essential element that is indispensable for life. The delicate physiological body iron balance is maintained by both systemic and cellular regulatory mechanisms. The iron-regulatory hormone hepcidin assures maintenance of adequate systemic iron levels and is regulated by circulating and stored iron levels, inflammation and erythropoiesis. The kidney has an important role in preventing iron loss from the body by means of reabsorption. Cellular iron levels are dependent on iron import, storage, utilization and export, which are mainly regulated by the iron response element-iron regulatory protein (IRE-IRP) system. In the kidney, iron transport mechanisms independent of the IRE-IRP system have been identified, suggesting additional mechanisms for iron handling in this organ. Yet, knowledge gaps on renal iron handling remain in terms of redundancy in transport mechanisms, the roles of the different tubular segments and related regulatory processes. Disturbances in cellular and systemic iron balance are recognized as causes and consequences of kidney injury. Consequently, iron metabolism has become a focus for novel therapeutic interventions for acute kidney injury and chronic kidney disease, which has fuelled interest in the molecular mechanisms of renal iron handling and renal injury, as well as the complex dynamics between systemic and local cellular iron regulation.
Collapse
|
40
|
Holden P, Nair LS. Deferoxamine: An Angiogenic and Antioxidant Molecule for Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:461-470. [PMID: 31184273 DOI: 10.1089/ten.teb.2019.0111] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Deferoxamine (DFO) has been in use for half a century as a Food and Drug Administration-approved iron chelator, but recent studies indicate a variety of properties that could expand this drug's application into the fields of tissue and regenerative engineering. DFO has been implicated as an angiogenic agent in studies on ischemia, wound healing, and bone regeneration because of its ability to upregulate hypoxia-inducible factor-1 alpha (HIF-1α) and other key downstream angiogenic factors. DFO has also demonstrated antioxidant capabilities unrelated to its iron-chelating properties, making it a potential modulator of the oxidative stress involved in the inflammation response. Together, these properties make DFO a potential bioactive molecule to promote wound healing and enhance tissue integration of biomaterials in vivo. Impact Statement Deferoxamine (DFO) is approved by the Food and Drug Administration as an iron chelator and is been used to treat iron overload. Recent studies indicate that DFO may have important applications in the growing field of tissue regeneration because of its unique properties of downregulating inflammation while promoting vascularization, thereby enhancing wound healing in vivo.
Collapse
Affiliation(s)
- Paige Holden
- The Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut.,Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| | - Lakshmi S Nair
- The Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut.,Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut.,Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, Connecticut.,Department of Material Science and Engineering, Institute of Material Science, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
41
|
Stamenkovic A, Pierce GN, Ravandi A. Phospholipid oxidation products in ferroptotic myocardial cell death. Am J Physiol Heart Circ Physiol 2019; 317:H156-H163. [DOI: 10.1152/ajpheart.00076.2019] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cell death is an important component of the pathophysiology of any disease. Myocardial disease is no exception. Understanding how and why cells die, particularly in the heart where cardiomyocyte regeneration is limited at best, becomes a critical area of study. Ferroptosis is a recently described form of nonapoptotic cell death. It is an iron-mediated form of cell death that occurs because of accumulation of lipid peroxidation products. Reactive oxygen species and iron-mediated phospholipid peroxidation is a hallmark of ferroptosis. To date, ferroptosis has been shown to be involved in cell death associated with Alzheimer’s disease, Huntington’s disease, cancer, Parkinson’s disease, and kidney degradation. Myocardial reperfusion injury is characterized by iron deposition as well as reactive oxygen species production. These conditions, therefore, favor the induction of ferroptosis. Currently there is no available treatment for reperfusion injury, which accounts for up to 50% of the final infarct size. This review will summarize the evidence that ferroptosis can induce cardiomyocyte death following reperfusion injury and the potential for this knowledge to open new therapeutic approaches for myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Aleksandra Stamenkovic
- Institute of Cardiovascular Sciences, Saint Boniface Hospital, and Departments of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Grant N. Pierce
- Institute of Cardiovascular Sciences, Saint Boniface Hospital, and Departments of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, Saint Boniface Hospital, and Departments of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
- Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
42
|
Otaki Y, Watanabe T, Takahashi H, Sugai T, Yokoyama M, Tamura H, Kato S, Nishiyama S, Arimoto T, Shishido T, Watanabe M. Impact of Iron Deficiency on Peripheral Artery Disease After Endovascular Therapy. Circ Rep 2019; 1:187-195. [PMID: 33693136 PMCID: PMC7889456 DOI: 10.1253/circrep.cr-18-0029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: Despite advances in endovascular therapy (EVT), peripheral artery disease (PAD) is a public health problem associated with high cardiovascular mortality. Iron deficiency (ID) is associated with poor clinical outcome in patients with heart disease, but whether ID is associated with the severity and clinical outcome of PAD remains unclear. Methods and Results: A total of 449 patients with PAD who received EVT and who had iron and red blood cell measurement were enrolled. ID was defined as transferrin saturation (TSAT) <20%, based on a previous report. TSAT and hemoglobin decreased with deteriorating Fontaine class. During a median follow-up period of 1,064 days, 71 major adverse cardiovascular and leg events (MACLE) and 47 major adverse cardiovascular events (MACE) were noted. All patients were divided into 2 groups based on the presence of ID. On Kaplan-Meier analysis, patients with ID had higher rates of MACE and MACLE than those without. On multivariate Cox proportional hazard regression analysis, TSAT and hemoglobin were independently associated with MACLE. Addition of TSAT to the known risk factors significantly improved the net reclassification index and integrated discrimination improvement. Conclusions: ID, as assessed by TSAT, was associated with the severity and clinical outcome of PAD, indicating that it could be a therapeutic target.
Collapse
Affiliation(s)
- Yoichiro Otaki
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Tetsu Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Hiroki Takahashi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Takayuki Sugai
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Miyuki Yokoyama
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Harutoshi Tamura
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Shigehiko Kato
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Satoshi Nishiyama
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Takanori Arimoto
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Tetsuro Shishido
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| | - Masafumi Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine Yamagata Japan
| |
Collapse
|
43
|
The uncertain effect of menopause on blood pressure. J Hum Hypertens 2019; 33:421-428. [PMID: 30899074 DOI: 10.1038/s41371-019-0194-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 01/10/2023]
Abstract
In affluent societies blood pressure increases with age from early life to the eighth decade with sex differences. Before middle age, lower blood pressure values are observed in women than in coeval men, whereas the reverse seems to occur thereafter. Menopause is considered the major determinant of blood pressure rise in women. If this hypothesis is well-founded, menopause can be regarded as one of the main cardiovascular risk factors, involving more than half of the human population, as well as the most ineluctable. In industrialized countries, age at menopause ranges between 50 and 52 years. The popular message is that fertile women are protected from cardiovascular risk by circulating estrogens, a privilege that is lost when postmenopausal women become not different from men from the point of view of risk factors and cardiovascular events. Nevertheless, the hypothesis that menopause or the estrogen decrease are per se associated to blood pressure increase is still under debate. Indeed, the epidemiological challenge is due to the coincidence between advancing menopause and aging, and also to the evidence that both menopause and blood pressure have common determinants such as body mass index, diet, smoking, and socio-economic class. The strongest doubt is whether menopause is a dependent or independent risk factor for high BP, i.e. whether its action on blood pressure-if any-is due directly to estrogen fall or to other indirect factors.
Collapse
|
44
|
Zhu Y, He B, Xiao Y, Chen Y. Iron metabolism and its association with dyslipidemia risk in children and adolescents: a cross-sectional study. Lipids Health Dis 2019; 18:50. [PMID: 30755213 PMCID: PMC6371579 DOI: 10.1186/s12944-019-0985-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 01/23/2019] [Indexed: 11/24/2022] Open
Abstract
Background Information on the association between iron metabolism and dyslipidaemia in children is limited. Thus, this study aims to evaluate the iron metabolic status of children with different body mass index (BMI) and to examine the association between iron metabolism and dyslipidaemia risk. Method In total, 1866 children and adolescents aged 7–18 were enrolled in this study, including 912 boys and 954 girls. In this cross-sectional study, parameters for anthropometry, lipids and iron metabolism including transferrin, soluble transferrin receptor (sTfR), ferritin and serum iron (SF) were evaluated. Data regarding demographic characteristics, diet, and physical activity were collected by self-reported questionnaires. Results The prevalence of dyslipidaemia and iron deficiency in children and adolescents increased based on BMI categories (both P < 0.05) and were 58.3 and 8.9% in subjects with obesity, respectively. The lowest SF and the highest ferritin levels were observed in subjects who were obese (both P < 0.001). Subjects with dyslipidaemia had lower SF, transferrin and sTfR levels by different BMI categories, and those who were obese had higher ferritin levels (all P < 0.05). Most importantly, higher concentrations of transferrin and sTfR were related to lower dyslipidaemia risk (OR for transferrin: 0.49, 95% CI: 0.33–0.71; OR for sTfR: 0.68, 95% CI: 0.46–0.99). Conclusions A downward trend in SF level by BMI categories and the highest ferritin level in subjects with obesity suggested that iron storage was associated with BMI in children and adolescents. Moreover, an inverse relationship was observed between transferrin and sTfR concentrations and dyslipidaemia risk in children with different BMI.
Collapse
Affiliation(s)
- Yanna Zhu
- Department of Maternal and Child Health, School of Public Health, and Global Health Institute (SGHI), Sun Yat-sen University, No.74 Zhongshan Road II, Guangzhou, 510080, Guangdong Province, China
| | - Baoting He
- Department of Maternal and Child Health, School of Public Health, and Global Health Institute (SGHI), Sun Yat-sen University, No.74 Zhongshan Road II, Guangzhou, 510080, Guangdong Province, China
| | - Yunjun Xiao
- Department of Nutrition and Food Hygiene, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yajun Chen
- Department of Maternal and Child Health, School of Public Health, and Global Health Institute (SGHI), Sun Yat-sen University, No.74 Zhongshan Road II, Guangzhou, 510080, Guangdong Province, China.
| |
Collapse
|
45
|
Kanamori Y, Murakami M, Sugiyama M, Hashimoto O, Matsui T, Funaba M. Hepcidin and IL-1β. VITAMINS AND HORMONES 2019; 110:143-156. [PMID: 30798809 DOI: 10.1016/bs.vh.2019.01.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hepcidin expression is determined through transcriptional regulation by systemic iron status. However, acute or chronic inflammation also increases the expression of hepcidin, which is associated with the dysregulation of iron metabolism in pathological conditions. Interleukin (IL)-6 has been suggested to be a principal molecule to confer inflammation-related hepcidin transcription, which is mediated via signal transducer and activator of transcription (STAT)-binding site on the hepcidin promoter. Recently, it has been uncovered that another pro-inflammatory cytokine IL-1β stimulates hepcidin expression through the distinct mechanism underlying IL-6-mediated hepcidin transcription. In addition to IL-6 induction, IL-1β stimulates expression of CCAAT-enhancer-binding protein (C/EBP)δ, a transcription factor, leading to transcriptional activation of hepcidin via C/EBP-binding site on the hepcidin promoter. Thus, hepcidin transcription is stimulated through multiple elements in response to proinflammatory cytokines. Relationships between increased production of IL-1β and dysregulated iron metabolism have been suggested in various diseases, which may be linked to overproduction of hepcidin.
Collapse
Affiliation(s)
- Yohei Kanamori
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Masaru Murakami
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, Japan
| | - Makoto Sugiyama
- Laboratory of Veterinary Anatomy, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Osamu Hashimoto
- Laboratory of Experimental Animal Science, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Tohru Matsui
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Masayuki Funaba
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan.
| |
Collapse
|
46
|
Ward LJ, Olausson P, Li W, Yuan XM. Proteomics and multivariate modelling reveal sex-specific alterations in distinct regions of human carotid atheroma. Biol Sex Differ 2018; 9:54. [PMID: 30594242 PMCID: PMC6311011 DOI: 10.1186/s13293-018-0217-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/12/2018] [Indexed: 01/05/2023] Open
Abstract
Background Atherosclerotic lesions are comprised of distinct regions with different proteomic profiles. Men and women develop differences in lesion phenotype, with lesions from women generally being more stable and less prone to rupture. We aimed to investigate the differences in proteomic profiles between sexes, including distinct lesion regions, to identify altered proteins that contribute to these differences observed clinically. Methods Carotid endarterectomy samples (ten men/ten women) were obtained, and intraplaque biopsies from three distinct regions (internal control, fatty streak and plaque) were analysed by tandem-mass spectrometry. Multivariate statistical modelling, using orthogonal partial least square-discriminant analysis, was used to discriminate the proteomes between men and women. Results Multivariate discriminant modelling revealed proteins from 16 functional groups that displayed sex-specific associations. Additional statistics revealed ten proteins that display region-specific alterations when comparing sexes, including proteins related to inflammatory response, response to reactive oxygen species, complement activation, transport and blood coagulation. Transport protein afamin and blood coagulation proteins antithrombin-III and coagulation factor XII were significantly increased in plaque region from women. Inflammatory response proteins lysozyme C and phospholipase A2 membrane-associated were significantly increased in plaque region from men. Limitations with this study are the small sample size, limited patient information and lack of complementary histology to control for cell type differences between sexes. Conclusions This pilot study, for the first time, utilises a multivariate proteomic approach to investigate sexual dimorphism in human atherosclerotic tissue, and provides an essential proteomic platform for further investigations to help understand sexual dimorphism and plaque vulnerability in atherosclerosis. Electronic supplementary material The online version of this article (10.1186/s13293-018-0217-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liam J Ward
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-581 85, Linköping, Sweden. .,Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Patrik Olausson
- Pain and Rehabilitation Centre, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Wei Li
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-581 85, Linköping, Sweden
| | - Xi-Ming Yuan
- Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
47
|
JNK facilitates IL-1β-induced hepcidin transcription via JunB activation. Cytokine 2018; 111:295-302. [DOI: 10.1016/j.cyto.2018.09.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/16/2018] [Accepted: 09/19/2018] [Indexed: 12/21/2022]
|
48
|
Hoda AI, Soha SZ, Manal ME, Mohamed RE, Samia AEE. New insight on premature atherosclerosis in Egyptian children with -thalassemia major. AFRICAN JOURNAL OF BIOCHEMISTRY RESEARCH 2018; 12:86-93. [DOI: 10.5897/ajbr2018.1005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
49
|
Poller WC, Pieber M, Boehm-Sturm P, Ramberger E, Karampelas V, Möller K, Schleicher M, Wiekhorst F, Löwa N, Wagner S, Schnorr J, Taupitz M, Stangl K, Stangl V, Ludwig A. Very small superparamagnetic iron oxide nanoparticles: Long-term fate and metabolic processing in atherosclerotic mice. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2575-2586. [PMID: 30179669 DOI: 10.1016/j.nano.2018.07.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/03/2018] [Accepted: 07/28/2018] [Indexed: 12/21/2022]
Abstract
We investigated the biotransformation of very small superparamagnetic iron oxide nanoparticles (VSOP) in atherosclerotic LDLR-/- mice. Transmission electron microscopy revealed an uptake of VSOP not only by macrophages but also by endothelial cells in liver, spleen, and atherosclerotic lesions and their accumulation in the lysosomal compartment. Using magnetic particle spectroscopy (MPS), we show that the majority of VSOP's superparamagnetic iron was degraded within 28 days. MPS spectrum shape indicated changes in the magnetic properties of VSOP during the biodegradation process. Experiments with primary murine bone marrow derived macrophages, primary murine liver sinusoidal endothelial cells, and primary human aortic endothelial cells demonstrated that loading with VSOP induced a differential response of cellular iron homeostasis mechanisms with increased levels of ferritin and iron transport proteins in macrophages and increased levels of ferritin in endothelial cells.
Collapse
Affiliation(s)
- Wolfram C Poller
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany.
| | - Melanie Pieber
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Berlin, Germany
| | - Philipp Boehm-Sturm
- Charité-Universitätsmedizin Berlin, Department of Experimental Neurology and Center for Stroke Research Berlin, Berlin, Germany; Charité-Universitätsmedizin Berlin, NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Berlin, Germany
| | - Evelyn Ramberger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Berlin, Germany
| | - Vasileios Karampelas
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Berlin, Germany
| | - Konstantin Möller
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Berlin, Germany
| | - Moritz Schleicher
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Berlin, Germany
| | | | - Norbert Löwa
- Physikalisch-Technische Bundesanstalt, Berlin, Germany
| | - Susanne Wagner
- Charité-Universitätsmedizin Berlin, Klinik für Radiologie, Berlin, Germany
| | - Jörg Schnorr
- Charité-Universitätsmedizin Berlin, Klinik für Radiologie, Berlin, Germany
| | - Matthias Taupitz
- Charité-Universitätsmedizin Berlin, Klinik für Radiologie, Berlin, Germany
| | - Karl Stangl
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Verena Stangl
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany.
| | - Antje Ludwig
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
50
|
Kell DB, Pretorius E. No effects without causes: the Iron Dysregulation and Dormant Microbes hypothesis for chronic, inflammatory diseases. Biol Rev Camb Philos Soc 2018; 93:1518-1557. [PMID: 29575574 PMCID: PMC6055827 DOI: 10.1111/brv.12407] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/11/2022]
Abstract
Since the successful conquest of many acute, communicable (infectious) diseases through the use of vaccines and antibiotics, the currently most prevalent diseases are chronic and progressive in nature, and are all accompanied by inflammation. These diseases include neurodegenerative (e.g. Alzheimer's, Parkinson's), vascular (e.g. atherosclerosis, pre-eclampsia, type 2 diabetes) and autoimmune (e.g. rheumatoid arthritis and multiple sclerosis) diseases that may appear to have little in common. In fact they all share significant features, in particular chronic inflammation and its attendant inflammatory cytokines. Such effects do not happen without underlying and initially 'external' causes, and it is of interest to seek these causes. Taking a systems approach, we argue that these causes include (i) stress-induced iron dysregulation, and (ii) its ability to awaken dormant, non-replicating microbes with which the host has become infected. Other external causes may be dietary. Such microbes are capable of shedding small, but functionally significant amounts of highly inflammagenic molecules such as lipopolysaccharide and lipoteichoic acid. Sequelae include significant coagulopathies, not least the recently discovered amyloidogenic clotting of blood, leading to cell death and the release of further inflammagens. The extensive evidence discussed here implies, as was found with ulcers, that almost all chronic, infectious diseases do in fact harbour a microbial component. What differs is simply the microbes and the anatomical location from and at which they exert damage. This analysis offers novel avenues for diagnosis and treatment.
Collapse
Affiliation(s)
- Douglas B. Kell
- School of ChemistryThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- The Manchester Institute of BiotechnologyThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| | - Etheresia Pretorius
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| |
Collapse
|