1
|
Zhang Y, Zheng L, Che T, Meng N. Dysregulation of Calcium Homeostasis: An Important Factor Leading to Ferroptosis in Cardiovascular Diseases. Cell Biol Int 2025; 49:589-605. [PMID: 40042118 DOI: 10.1002/cbin.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/31/2024] [Accepted: 02/16/2025] [Indexed: 05/14/2025]
Abstract
Cardiovascular disease is a circulatory system disease involving the heart and blood vessels, which is one of the main causes of human health loss and even life-threatening. Ca2+ is an important signal molecule. Free calcium ions in the cytoplasm are involved in various physiological and biochemical reactions of cells. Ferroptosis is a programmed cell death driven by lipid peroxidation dependent on free ferrous ions. The essence of ferroptosis is the accumulation of lipid peroxide caused by the increase of intracellular ferrous ion content, which leads to the damage of the phospholipid membrane and eventually cell death. Studies have shown that calcium homeostasis and ferroptosis are involved in the occurrence and development of cardiovascular diseases, but the relationship between them remains to be clarified. This article reviews the various pathways regulating calcium homeostasis in cells and the occurrence and development mechanism of ferroptosis, and discusses the relationship between the two in cardiovascular diseases, which is expected to provide novel and important strategies for alleviating and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Yifan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Leiyin Zheng
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Tongtong Che
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan, China
| |
Collapse
|
2
|
Kumfu S, Sripetchwandee J, Thonusin C, Maneechote C, Arunsak B, Chunchai T, Kongkaew A, Chattipakorn SC, Chattipakorn N. Mitochondrial dynamic modulators attenuate iron overload-mediated cardiac toxicity via decreased mitochondrial fission, mitophagy/autophagy, and apoptosis in iron-overloaded rats. Arch Biochem Biophys 2025; 767:110354. [PMID: 39988036 DOI: 10.1016/j.abb.2025.110354] [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: 09/18/2024] [Revised: 01/12/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
One of the leading causes of death for individuals with iron overload is iron overload cardiomyopathy (IOC). Iron overload causes cardiac mitochondrial dysfunction, which ultimately results in heart failure and death. The potential mechanism of iron overload-induced mitochondrial dysfunction involves the disequilibrium between cardiac mitochondrial fission and fusion. Nevertheless, the information regarding cardiac mitochondrial dynamics under iron overload conditions remains limited. The roles of mitochondrial dynamics were identified in IOC. To induce iron overload, male Wistar rats were injected with iron dextran for four weeks. Then, while continuing iron dextran injection, four groups of iron-overloaded rats were given injections of either vehicle, mitochondrial fusion promoter (M1), mitochondrial division inhibitor 1 (Mdivi-1), or iron chelator deferoxamine (DFO) for two weeks. In the non-iron loaded (control) group, rats received vehicles without iron dextran injection. Cardiac function, mitochondrial function, mitochondrial dynamics, mitophagy/autophagy, and apoptosis were assessed at the end of treatment. The increased expression of mitochondrial fission-, mitophagy/autophagy-, and apoptosis-related proteins were correlated with impaired mitochondrial and cardiac functions in iron-overloaded rats. Interestingly, both mitochondrial dynamics modulators reduced cardiac mitochondrial fission, mitophagy/autophagy, and apoptosis, as well as restored cardiac function to be comparable to those treated with iron chelator DFO. Our findings indicated that the imbalance of mitochondrial dynamics is a potential mechanism responsible for cardiomyocyte death induced by IOC, and this could be a novel target for interventions for IOC via either the promotion of mitochondrial fusion or the inhibition of mitochondrial fission.
Collapse
Affiliation(s)
- Sirinart Kumfu
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Jirapas Sripetchwandee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Chanisa Thonusin
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
3
|
Maneechote C, Khuanjing T, Ongnok B, Arinno A, Prathumsap N, Chunchai T, Arunsak B, Nawara W, Chattipakorn SC, Chattipakorn N. Targeting mitochondrial dynamics emerges as an effective strategy of cardioprotection against trastuzumab-induced mitochondrial functional aberrations and cardiotoxicity in rats. Eur J Pharmacol 2025; 999:177685. [PMID: 40288556 DOI: 10.1016/j.ejphar.2025.177685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Trastuzumab (Trz) is a targeted anticancer therapy that specifically acts on tumors overexpressing the human epidermal growth factor receptor 2 (HER2) protein. Previous research has shown that Trz can induce cardiotoxicity by altering mitochondrial function. While modulating mitochondrial dynamics with Mdivi-1 and M1 has shown cardioprotective effects in various cardiac conditions, their impact on Trz-induced cardiotoxicity in rats remains unclear. In this study, thirty-two male Wistar rats were divided into a control group (CON, n = 8) and a Trz-treated group (4 mg/kg/day, i.p. for 7 days, n = 24). The Trz group was further randomized into subgroups receiving either: 1) vehicle (VEH, 3 % DMSO, i.p., n = 8), 2) the mitochondrial fission inhibitor Mdivi-1 (MDV, 1.2 mg/kg/day, i.p., n = 8), or 3) the mitochondrial fusion promoter M1 (2 mg/kg/day, i.p., n = 8). All interventions began on the first day of Trz administration and continued for 7 days. At the end, cardiac function was then assessed, and heart tissue was collected for biochemical analysis. Trz-treated rats exhibited cardiotoxicity, including cardiac dysfunction and injury, as well as disrupted mitochondrial and autophagic processes, increased inflammation, oxidative stress, apoptosis, ferroptosis, and pyroptosis. Co-administration of either Mdivi-1 or M1 with Trz alleviated these harmful effects, suggesting that modulating mitochondrial dynamics might offer a novel therapeutic strategy to mitigate Trz-induced cardiotoxicity.
Collapse
Affiliation(s)
- Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Benjamin Ongnok
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Apiwan Arinno
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nanthip Prathumsap
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Wichwara Nawara
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
4
|
Bazi A, Khanahmad A, Khazaee-Nasirabadi MH, Pirouzbakht M, Ghorbani Biregani K, Peymaninezhad F, Mirzaee Khalilabadi R. Long-Term and Transient Calcium Channel Blockers; A Systematic Review of Their Role in the Management of Cardiomyopathy in Transfusion-Dependent Thalassemia. Hemoglobin 2025; 49:111-125. [PMID: 40069102 DOI: 10.1080/03630269.2025.2470718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/12/2024] [Accepted: 02/06/2025] [Indexed: 04/22/2025]
Abstract
Calcium channel blockers (CCBs) for long-term (L) and transient (T) calcium channels (LTCC and TTCC) on cardiomyocytes have been suggested to manage iron-induced cardiomyopathy in transfusion-dependent thalassemia patients. However, the results of clinical trials on the effectiveness of CCBs have been conflicting. Here, we systematically reviewed previous studies to investigate the potential factors that could act as therapeutic modifiers and explain these discrepancies. This systematic review was conducted employing the PRISMA guideline to retrieve clinical trials and animal studies investigating the efficacy of CCBs. Studies in the following databases were collected: Web of Science, PubMed, Scopus, Google Scholar, Clinical Trials, Iranian Registry for Clinical Trials, and Cochrane CENTRAL. Keywords included the trade and generic names of various CCBs, thalassemia, and cardiomyopathy. Our Primary search resulted in 297 studies, of which 21 (n = 7 trials and n = 14 animal studies) were further analyzed. The most important parameters that could potentially influence the clinical effectiveness of CCBs in managing iron-induced cardiomyopathy included baseline cardiac iron content, diversity of iron entry routes (LTCCs, TTCCs, DMT-1, etc.), type of CCBs used, iron-induced irreversible functional/structural cardiac changes, iron-Ca2+ joint metabolic dysregulation, deregulated expression of LTCCs and TTCCs, interaction of CCBs with iron chelators, disease-related complications, interactions of CCBs with various supplements used by patients, vitamin D and other nutrient deficiencies, and duration of treatment with CCBs. These items should be considered in future trials to draw more robust conclusions about the effectiveness of CCBs in preventing cardiac iron deposition and associated cardiomyopathy in TDT patients.
Collapse
Affiliation(s)
- Ali Bazi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Khanahmad
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hossein Khazaee-Nasirabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Pirouzbakht
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Kobra Ghorbani Biregani
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Peymaninezhad
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Roohollah Mirzaee Khalilabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
5
|
Aiemongkot S, Ruschadaariyachat S, Changsangfa C, Nuamsee K, Viwatpinyo K, Chaichompoo P, Svasti S, Buranaamnuay K. Male reproductive phenotype alterations in heterozygous β-globin gene knockout thalassemia (BKO) mice as a model for β-thalassemia patients. Sci Rep 2025; 15:4903. [PMID: 39929883 PMCID: PMC11811285 DOI: 10.1038/s41598-025-87619-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
Heterozygous β-globin gene knockout thalassemia (BKO) mice derived from C57BL/6 wild-type (WT) mice have phenotypic of β-thalassemia (BT) and have been widely used for studying this disease except reproductive disorders. The present study determined whether male BKO mice recapitulate reproductive problems as BT men. Mice were randomly assigned into groups depending on the genotype (WT vs. BKO) and intervention (control vs. iron-loaded). Euthanized mice were collected blood, testes, epididymides, hypothalamus, and anterior pituitary for assessing hematological parameters, plasma iron and testosterone levels, testis iron levels, sperm characteristics, and histological alterations. Iron administration caused significant increases of plasma and testis iron levels (p < 0.001) but had no significant influence on the hematological profile of BKO mice, which indeed had fewer erythrocyte, hemoglobin, and hematocrit but had greater reticulocyte than WT (p < 0.001 to p = 0.017). Furthermore, irrespective of the genotype, iron administration decreased plasma testosterone levels (p = 0.03 to p > 0.05), total sperm count (p < 0.001), and percent normal sperm morphology (p ≤ 0.01). Based on Perls' Prussian blue staining, excess iron was ubiquitously present in the anterior pituitary and testicular interstitium of iron-loaded mice. This mineral, however, caused no significant changes in reproductive organs microstructure as visualized by hematoxylin and eosin staining. In conclusion, besides physiological dysfunction of many organ systems, iron-loaded male BKO mice exhibit reproductive problems and abnormal sperm characteristics similar to BT men. Therefore, this animal model seems invaluable for future biomedical research involved in various aspects of BT-related male reproductive disorders.
Collapse
Affiliation(s)
- Suparada Aiemongkot
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Sukanya Ruschadaariyachat
- Office of Research and Innovation Affair, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Chinarat Changsangfa
- Office of Research and Innovation Affair, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Khanita Nuamsee
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
- Department of Biochemistry, Faculty of Medicine, Bangkokthonburi University, Bangkok, Thailand
| | - Kittikun Viwatpinyo
- Dapartment of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
| | - Pornthip Chaichompoo
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Kakanang Buranaamnuay
- Molecular Biosciences Cluster, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
6
|
Wang Z, Wu C, Yin D, Dou K. Ferroptosis: mechanism and role in diabetes-related cardiovascular diseases. Cardiovasc Diabetol 2025; 24:60. [PMID: 39920799 PMCID: PMC11806630 DOI: 10.1186/s12933-025-02614-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
Cardiovascular diseases represent the principal cause of death and comorbidity among people with diabetes. Ferroptosis, an iron-dependent non-apoptotic regulated cellular death characterized by lipid peroxidation, is involved in the pathogenesis of diabetic cardiovascular diseases. The susceptibility to ferroptosis in diabetic hearts is possibly related to myocardial iron accumulation, abnormal lipid metabolism and excess oxidative stress under hyperglycemia conditions. Accumulating evidence suggests ferroptosis can be the therapeutic target for diabetic cardiovascular diseases. This review summarizes ferroptosis-related mechanisms in the pathogenesis of diabetic cardiovascular diseases and novel therapeutic choices targeting ferroptosis-related pathways. Further study on ferroptosis-mediated cardiac injury can enhance our understanding of the pathophysiology of diabetic cardiovascular diseases and provide more potential therapeutic choices.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Wu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Yin
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Kefei Dou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
7
|
Qin S, Zhu C, Chen C, Sheng Z, Cao Y. An emerging double‑edged sword role of ferroptosis in cardiovascular disease (Review). Int J Mol Med 2025; 55:16. [PMID: 39540363 PMCID: PMC11573318 DOI: 10.3892/ijmm.2024.5457] [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: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The pathophysiology of cardiovascular disease (CVD) is complex and presents a serious threat to human health. Cardiomyocyte loss serves a pivotal role in both the onset and progression of CVD. Among various forms of programmed cell death, ferroptosis, along with apoptosis, autophagy and pyroptosis, is closely linked to the advancement of CVD. Ferroptosis, a mechanism of cell death, is driven by the buildup of oxidized lipids and excess iron. This pathway is modulated by lipid, amino acid and iron metabolism. Key characteristics of ferroptosis include disrupted iron homeostasis, increased peroxidation of polyunsaturated fatty acids due to reactive oxygen species, decreased glutathione levels and inactivation of glutathione peroxidase 4. Treatments targeting ferroptosis could potentially prevent or alleviate CVD by inhibiting the ferroptosis pathway. Ferroptosis is integral to the pathogenesis of several types of CVD and inhibiting its occurrence in cardiomyocytes could be a promising therapeutic strategy for the future treatment of CVD. The present review provided an in‑depth analysis of advancements in understanding the mechanisms underlying ferroptosis. The present manuscript summarized the interplay between ferroptosis and CVDs, highlighting its dual roles in these conditions. Additionally, potential therapeutic targets within the ferroptosis pathway were discussed, alongside the current limitations and future directions of these novel treatment strategies. The present review may offer novel insights into preventive and therapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Sirun Qin
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Can Zhu
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Chenyang Chen
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhe Sheng
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yu Cao
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
8
|
Satish V, Maliha M, Chi KY, Kharawala A, Seo J, Apple S, Alhuarrat MAD, Palaiodimos L, Di Biase L, Krumerman A, Ferrick K. Catheter Ablation of Atrial Fibrillation in Infiltrative Cardiomyopathies: A Narrative Review. J Cardiovasc Electrophysiol 2025; 36:276-285. [PMID: 39506617 DOI: 10.1111/jce.16487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/09/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024]
Abstract
Atrial and ventricular arrhythmias are common in patients with Infiltrative heart diseases. This review discusses ablative techniques for arrhythmias in amyloidosis, sarcoidosis, hemochromatosis, and glycogen storage disorders, primarily focusing on atrial fibrillation (AF). A thorough literature review was conducted on the MEDLINE database to synthesize current knowledge and propose future research directions. AF is the most common arrhythmia identified in patients with amyloidosis due to cellular infiltration and atrial dilation. While catheter ablation is associated with a significantly lower rate of all-cause mortality and admission rate, conflicting data exist regarding the higher risk of pericardial effusion, in-hospital mortality, length of stay, and cost of hospitalization. Cardiac sarcoid predisposes AF due to granulomas, atrial dilation, and scarring. Studies demonstrate encouraging outcomes and low recurrence rates in these patients who undergo ablation for AF, with no difference in complications compared to those without sarcoidosis. AF is the most common arrhythmia in hereditary hemochromatosis (HH), secondary to increased myocardial iron stores and elevated oxidative stress, and is primarily managed by chelation. Scant reports regarding ablation are described for HH and glycogen storage disorders. Catheter ablation is a safe and effective modality for the treatment of AF in infiltrative cardiomyopathy. Future large-scale trials are needed to confirm these findings.
Collapse
Affiliation(s)
- Vikyath Satish
- Department of Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Maisha Maliha
- Department of Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kuan-Yu Chi
- Department of Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Amrin Kharawala
- Department of Medicine, Division of Cardiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jiyoung Seo
- Department of Medicine, Division of Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Samuel Apple
- Department of Medicine, Division of Cardiology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Majd Al Deen Alhuarrat
- Department of Medicine, Division of Cardiology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Leonidas Palaiodimos
- Department of Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Luigi Di Biase
- Department of Medicine, Division of Cardiology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrew Krumerman
- Department of Medicine, Division of Cardiology, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Kevin Ferrick
- Department of Medicine, Division of Cardiology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
9
|
Zhang S, Wang D, Ding Y, Li Y, Wang Y, Zeng J. Inhibition of calpain reduces oxidative stress and attenuates pyroptosis and ferroptosis in Clostridium perfringens Beta-1 toxin-induced macrophages. Microbiol Res 2024; 289:127916. [PMID: 39342748 DOI: 10.1016/j.micres.2024.127916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/21/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
Clostridium perfringens Beta-1 toxin (CPB1) is a lethal toxin, which can lead to necrotic enteritis, but the pathological mechanism has not been elucidated. We investigated whether reactive oxygen species (ROS) participated in CPB1-induced pyroptosis and ferroptosis, and investigated the effects of calpain on CPB1-induced oxidative stress and inflammation. Scavenging ROS by N-Acetyl-L cysteine (NAC) led to the reduction of ROS, inhibited the death of macrophages, cytoplasmic swelling and membrane rupture, the expression of pyroptosis-related proteins and proinflammatory factor, while increased the expression of anti-inflammatory factors in cells treated with rCPB1. Adenosine triphosphate (ATP) synthase, H+ transporting, mitochondrial F1 complex, alpha subunit 1 (ATP5A1) was identified specifically interact with rCPB1. Silencing ATP5A1 inhibited accumulation of ATP and ROS, leaded to less cytoplasmic swelling and membrane rupture, attenuated pyroptosis and inflammation in rCPB1-treated cells. We also found that rCPB1 induces ferroptosis in macrophages, and the level of ferroptosis was similar with H2O2. Of note, H2O2 is a major ROS source, indicated that ROS production may play a major role in the regulation of ferroptosis in macrophages treated with rCPB1. This finding was further corroborated in rCPB1- induced human acute monocytic leukemia cells, which were treated with NAC. In addition, the inhibition of ferroptosis using liproxstatin-1 inhibited the shriveled mitochondrial morphology, increased the expression of glutathione peroxidase 4, nicotinamide adenine dinucleotide (phosphate) hydrogen: quinone oxidoreductase 1 and cysteine/glutamic acid reverse transport solute carrier family 7 members 11, decreased the expression of heme oxygenase 1, nuclear receptor coactivator 4 and transferrin receptor proteins, reduced malondialdehyde and lipid peroxidation levels, and increased intracellular L-glutathione levels in cells treated with rCPB1. Furthermore, calpain inhibitor PD151746 was used to investigate how pyroptosis and ferroptosis were involved simultaneously in rCPB1-treated macrophages. We showed that PD151746 inhibited ATP and ROS production, reversed the representative pyroptosis/ferroptosis indicators and subsequently reduced inflammation. The above findings indicate that rCPB1 might lead to macrophage pyroptosis and ferroptosis through the large and sustained increase in intracellular calpain and oxidative stress, further lead to inflammation.
Collapse
Affiliation(s)
- Siyu Zhang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan 750021, China
| | - Dong Wang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan 750021, China
| | - Yawen Ding
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan 750021, China
| | - Yong Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan 750021, China
| | - Yujiong Wang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan 750021, China.
| | - Jin Zeng
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
10
|
Tian M, Huang X, Li M, Lou P, Ma H, Jiang X, Zhou Y, Liu Y. Ferroptosis in diabetic cardiomyopathy: from its mechanisms to therapeutic strategies. Front Endocrinol (Lausanne) 2024; 15:1421838. [PMID: 39588340 PMCID: PMC11586197 DOI: 10.3389/fendo.2024.1421838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is defined as structural and functional cardiac abnormalities in diabetes, and cardiomyocyte death is the terminal event of DCM. Ferroptosis is iron-dependent oxidative cell death. Evidence has indicated that iron overload and ferroptosis play important roles in the pathogenesis of DCM. Mitochondria, an important organelle in iron homeostasis and ROS production, play a crucial role in cardiomyocyte ferroptosis in diabetes. Studies have shown some anti-diabetic medicines, plant extracts, and ferroptosis inhibitors might improve DCM by alleviating ferroptosis. In this review, we systematically reviewed the evidence of ferroptosis in DCM. Anti-ferroptosis might be a promising therapeutic strategy for the treatment of DCM.
Collapse
Affiliation(s)
- Meimei Tian
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinli Huang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Min Li
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pingping Lou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huijie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Xinli Jiang
- Department of Ophthalmology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yaru Zhou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yan Liu
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
11
|
Vera-Aviles M, Kabir SN, Shah A, Polzella P, Lim DY, Buckley P, Ball C, Swinkels D, Matlung H, Blans C, Holdship P, Nugent J, Anderson E, Desborough M, Piechnik S, Ferreira V, Lakhal-Littleton S. Intravenous iron therapy results in rapid and sustained rise in myocardial iron content through a novel pathway. Eur Heart J 2024; 45:4497-4508. [PMID: 38917062 PMCID: PMC11544312 DOI: 10.1093/eurheartj/ehae359] [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: 01/12/2024] [Revised: 05/10/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND AND AIMS Intravenous iron therapies contain iron-carbohydrate complexes, designed to ensure iron becomes bioavailable via the intermediary of spleen and liver reticuloendothelial macrophages. How other tissues obtain and handle this iron remains unknown. This study addresses this question in the context of the heart. METHODS A prospective observational study was conducted in 12 patients receiving ferric carboxymaltose (FCM) for iron deficiency. Myocardial, spleen, and liver magnetic resonance relaxation times and plasma iron markers were collected longitudinally. To examine the handling of iron taken up by the myocardium, intracellular labile iron pool (LIP) was imaged in FCM-treated mice and cells. RESULTS In patients, myocardial relaxation time T1 dropped maximally 3 h post-FCM, remaining low 42 days later, while splenic T1 dropped maximally at 14 days, recovering by 42 days. In plasma, non-transferrin-bound iron (NTBI) peaked at 3 h, while ferritin peaked at 14 days. Changes in liver T1 diverged among patients. In mice, myocardial LIP rose 1 h and remained elevated 42 days after FCM. In cardiomyocytes, FCM exposure raised LIP rapidly. This was prevented by inhibitors of NTBI transporters T-type and L-type calcium channels and divalent metal transporter 1. CONCLUSIONS Intravenous iron therapy with FCM delivers iron to the myocardium rapidly through NTBI transporters, independently of reticuloendothelial macrophages. This iron remains labile for weeks, reflecting the myocardium's limited iron storage capacity. These findings challenge current notions of how the heart obtains iron from these therapies and highlight the potential for long-term dosing to cause cumulative iron build-up in the heart.
Collapse
Affiliation(s)
- Mayra Vera-Aviles
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Syeeda Nashitha Kabir
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Akshay Shah
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Paolo Polzella
- Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Dillon Yee Lim
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Poppy Buckley
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Charlotte Ball
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Dorine Swinkels
- Department of Laboratory Medicine, Iron Expertise Centre, Radboud University Medical Centre, Nijmegen, The Netherlands
- Iron Expertise Centre, Sanquin Blood Bank, Amsterdam, The Netherlands
| | - Hanke Matlung
- Sanquin Diagnostic Services, Amsterdam, The Netherlands
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Colin Blans
- Sanquin Diagnostic Services, Amsterdam, The Netherlands
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Philip Holdship
- Department of Earth Sciences, University of Oxford, Oxford, United Kingdom
| | - Jeremy Nugent
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, United Kingdom
| | - Edward Anderson
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, United Kingdom
| | - Michael Desborough
- Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Stefan Piechnik
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, Oxford, United Kingdom
| | - Vanessa Ferreira
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, Oxford, United Kingdom
| | - Samira Lakhal-Littleton
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, United Kingdom
| |
Collapse
|
12
|
Zheng L, Ding Y, Fang S, Yang W, Chen J, Ma J, Wang M, Wang J, Zhang F, Guo X, Zhang K, Shu GF, Weng Q, Wu F, Zhao Z, Chen M, Jiansong J. Potentiated Calcium Carbonate with Enhanced Calcium Overload Induction and Acid Neutralization Capabilities to Boost Chemoimmunotherapy against Liver Cancer. ACS NANO 2024; 18:27597-27616. [PMID: 39342637 DOI: 10.1021/acsnano.4c08690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Unfavorable phenotypes characterized by low immunogenicity and acidity within the tumor microenvironment (TME) contribute to immunosuppression and therapeutic resistance. Herein, we rationally synthesized a multifunctional nanoregulator by encapsulating DOX and erianin into calcium carbonate (CaCO3)-based nanoparticles using a modified double emulsion method. The DOX and erianin-loaded CaCO3-based nanoparticles, termed DECaNPs, could effectively induce the calcium overload by triggering calcium influx and absorbing CaCO3 nanoparticles. Additionally, DECaNPs also neutralize the acidic TME by interacting with extracellular protons and limiting lactic acid production, a result of metabolic remodeling in cancer cells. As a result, DECaNPs elicit cellular oxidative stress damage, which mediates the activation of ferroptosis/apoptosis hybrid pathways, and profound immunogenic cell death. Treatment with DECaNPs could inhibit the growth of tumors by promoting oxidative stress, acid neutralization, metabolic remodeling, and protective antitumor immunity in vivo. In addition, DECaNPs could synergistically amplify the antitumor effects of αPD-L1 in a bilateral tumor model by eliciting systemic immune responses. In all, our work presents the preparation of CaCO3-based nanoregulators designed to reverse the unfavorable TME and enhance αPD-L1 immunotherapy through multiple mechanisms.
Collapse
Affiliation(s)
- Liyun Zheng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Yiming Ding
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Wenjing Yang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Jiale Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Ji Ma
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Mengyuan Wang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jiaoli Wang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Feng Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Xiaoju Guo
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Kun Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Gao-Feng Shu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Qiaoyou Weng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Fazong Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Ji Jiansong
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, China
| |
Collapse
|
13
|
Zeng H, Xu J, Wu R, Wang X, Jiang Y, Wang Q, Guo J, Xiao F. FTO alleviated ferroptosis in septic cardiomyopathy via mediating the m6A modification of BACH1. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167307. [PMID: 38897256 DOI: 10.1016/j.bbadis.2024.167307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/28/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
Sepsis is a global health challenge that results in systemic inflammation, oxidative stress, and multi-organ dysfunction, with the heart being particularly susceptible. This study aimed to elucidate the effect of FTO, a key regulator in m6A methylation in septic cardiomyopathy, and its potential therapeutic implications. Cellular and animal models of septic myocardial injury were established. Moreover, it was revealed that ferroptosis, which is a form of programmed necrosis occurring with iron dependence, was activated within cardiomyocytes during septic conditions. The overexpression of FTO-suppressed ferroptosis alleviated heart inflammation and dysfunction and improved survival rates in vivo. However, the protective effects of FTO were attenuated by the overexpression of BACH1, which is a molecule negatively correlated with FTO. Mechanistically, FTO modulated the m6A modification of BACH1, suggesting a complex interplay in the regulation of cardiomyocyte damage and sepsis. Our findings reveal the potential of targeting the FTO/BACH1 axis and ferroptosis inhibitors as therapeutic strategies for sepsis-induced cardiac injuries.
Collapse
Affiliation(s)
- Hua Zeng
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Junmei Xu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Rui Wu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xin Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yaqing Jiang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qing Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Jiali Guo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Feng Xiao
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
14
|
Yu X, Li M, Chen B, Qi Y, Guan X. Association between serum iron levels and atherosclerotic cardiovascular diseases among American older adults: a cross-sectional study based on the National Health and Nutrition Examination Survey, 2009-2018. Front Nutr 2024; 11:1457236. [PMID: 39385780 PMCID: PMC11463155 DOI: 10.3389/fnut.2024.1457236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
Background There is controversy regarding the relationship between serum iron levels and atherosclerotic cardiovascular disease (ASCVD). Objective To investigate the relationship between serum iron levels and ASCVD among older adults using data from the 2009-2018 National Health and Nutrition Examination Survey (NHANES). Methods We performed a cross-sectional analysis involving 8,682 participants aged 60 years and older, with complete data on serum iron levels and confirmed ASCVD status, sourced from the 2009-2018 National Health and Nutrition Examination Survey (NHANES). Multivariable logistic regression models were used to examine the association between serum iron levels and ASCVD. To assess the consistency of this association across different demographic groups, subgroup analyses, and interaction tests were performed. Results The group with the highest serum iron levels (fourth quartile, 100-369 μg/dL) exhibited several distinct characteristics: they were the youngest on average (69.57 ± 6.91 years), had the highest proportion of males (61.42%), and the highest hemoglobin levels (14.43 ± 1.33 g/dL). This group also showed the lowest iron supplement usage (19.71 ± 12.85 mg/30 days), white blood cell counts (6.73 ± 2.41 1,000 cells/μL), and serum creatinine levels (0.98 ± 0.45 mg/dL). Moreover, they had higher levels of education and income, a higher likelihood of being married, and a lower body mass index (BMI). Additionally, they had significantly lower rates of diabetes, hypertension, stroke, and heart attacks (all p < 0.05). After adjusting for potential confounders, a linear relationship between serum iron levels and ASCVD was initially observed (OR = 0.97; 95% CI, 0.95-0.99, p < 0.05). However, further analysis using a two-part logistic regression model with an inflection point at 131 μg/dL revealed more nuanced results. For serum iron levels below 131 μg/dL, each 10 μg/dL increase was associated with a 4% decrease in the odds of ASCVD (OR = 0.96; 95% CI, 0.93-0.98, p < 0.001). Conversely, for serum iron levels above 131 μg/dL, each 10 μg/dL increase corresponded to a 1% increase in the odds of ASCVD, though this finding was not statistically significant (OR = 1.01; 95% CI, 0.98-1.08, p > 0.05). Conclusion In the US elderly population, serum iron levels are negatively associated with ASCVD, particularly when serum iron levels are below 131 μg/dL.
Collapse
Affiliation(s)
| | | | | | | | - Xiuru Guan
- Department of Laboratory Diagnostics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
15
|
Ye H, Wu L, Liu Y. Iron metabolism in doxorubicin-induced cardiotoxicity: From mechanisms to therapies. Int J Biochem Cell Biol 2024; 174:106632. [PMID: 39053765 DOI: 10.1016/j.biocel.2024.106632] [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: 05/28/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Doxorubicin (DOX) is an anti-tumor agent for chemotherapy, but its use is often hindered by the severe and life-threatening side effect of cardiovascular toxicity. In recent years, studies have focused on dysregulated iron metabolism and ferroptosis, a unique type of cell death induced by iron overload, as key players driving the development of DOX-induced cardiotoxicity (DIC). Recent advances have demonstrated that DOX disturbs normal cellular iron metabolism, resulting in excessive iron accumulation and ferroptosis in cardiomyocytes. This review will explore how dysregulated iron homeostasis and ferroptosis drive the progression of DIC. We will also discuss the current approaches to target iron metabolism and ferroptosis to mitigate DIC. Besides, we will discuss the limitations and challenges for clinical translation for these therapeutic regimens.
Collapse
Affiliation(s)
- Hua Ye
- Department of Burns & Plastic and Wound Repair, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China.
| | - Lin Wu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yanmei Liu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
16
|
Meloni A, Pistoia L, Ricchi P, Longo F, Cecinati V, Sorrentino F, Borsellino Z, Bagnato S, Rossi V, Fina P, Riva A, Renne S, Peritore G, Positano V, Cademartiri F. Magnetic Resonance Evaluation of Tissue Iron Deposition and Cardiac Function in Adult Regularly Transfused Thalassemia Intermedia Compared with Thalassemia Major Patients. J Clin Med 2024; 13:4791. [PMID: 39200932 PMCID: PMC11355279 DOI: 10.3390/jcm13164791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Objectives: This multicenter, retrospective, population-based, matched-cohort study compared clinical characteristics and magnetic resonance imaging (MRI) findings, including hepatic, pancreatic, and cardiac iron levels and cardiac function, between 135 adult regularly transfused thalassemia intermedia (TI) patients (44.73 ± 12.16 years, 77 females) and 135 age- and sex-matched thalassemia major (TM) patients (43.35 ± 9.83 years, 77 females), enrolled in the Extension-Myocardial Iron Overload in Thalassemia Network. Methods: The MRI protocol included the quantification of hepatic, pancreatic, and cardiac iron levels (R2* technique), the assessment of biventricular function parameters (cine images), and the detection of replacement myocardial fibrosis (late gadolinium enhancement technique). Results: Age, sex, frequency of splenectomy and chelation, and serum ferritin levels were not significantly different (p > 0.05) between the two groups, but TI patients started regular transfusions significantly later (p < 0.0001) and showed significantly lower pre-transfusion hemoglobin levels (p = 0.005). No difference was found in hepatic iron levels (p = 0.853). TI patients exhibited significantly lower pancreatic R2* values (p < 0.0001), also correcting for the duration of regular transfusions, and significantly lower cardiac R2* values (p < 0.0001). In the receiver operating characteristic analysis, pancreatic iron was the strongest discriminator between the two diseases. Left and right ventricular end-diastolic volume indexes were significantly higher in TI than in TM patients (p = 0.003 and p = 0.046, respectively), but the correction for the duration of regular transfusions removed the disease-specific differences (p > 0.05). Left ventricular (LV) mass index was significantly higher in TI (p = 0.049), while no difference (p > 0.05) was found in biventricular ejection fractions and replacement myocardial fibrosis. Conclusions: TI patients showed lower pancreatic and cardiac iron burden and more pronounced LV hypertrophy. These differences could not be explained by the different duration of the transfusional regimen.
Collapse
Affiliation(s)
- Antonella Meloni
- Bioengineering Unit, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.)
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | - Laura Pistoia
- Bioengineering Unit, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.)
- Unità Operativa Complessa Ricerca Clinica, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | - Paolo Ricchi
- Unità Operativa Semplice Dipartimentale Malattie Rare del Globulo Rosso, Azienda Ospedaliera di Rilievo Nazionale “A. Cardarelli”, 80131 Napoli, Italy;
| | - Filomena Longo
- Unità Operativa Day Hospital della Talassemia e delle Emoglobinopatie, Azienda Ospedaliero-Universitaria “S. Anna”, 44124 Cona (FE), Italy;
| | - Valerio Cecinati
- Struttura Semplice di Microcitemia, Ospedale “SS. Annunziata” ASL Taranto, 74123 Taranto, Italy;
| | | | - Zelia Borsellino
- Unità Operativa Complessa Ematologia con Talassemia, “ARNAS” Civico Di Cristina Benfratelli, 90134 Palermo, Italy;
| | - Sergio Bagnato
- Ematologia Microcitemia, Ospedale San Giovanni di Dio—ASP Crotone, 88900 Crotone, Italy;
| | - Vincenza Rossi
- Unità Operativa Complessa Ematologia, Ospedale di Cosenza, 87100 Cosenza, Italy;
| | - Priscilla Fina
- Unità Operativa Complessa Diagnostica per Immagini, Ospedale “Sandro Pertini”, 00157 Roma, Italy;
| | - Ada Riva
- Struttura Complessa di Radiologia, Ospedale “SS. Annunziata” ASL Taranto, 74100 Taranto, Italy;
| | - Stefania Renne
- Struttura Complessa di Cardioradiologia-UTIC, Presidio Ospedaliero “Giovanni Paolo II”, 88046 Lamezia Terme (CZ), Italy;
| | - Giuseppe Peritore
- Unità Operativa Complessa di Radiologia, “ARNAS” Civico Di Cristina Benfratelli, 90127 Palermo, Italy;
| | - Vincenzo Positano
- Bioengineering Unit, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.)
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | - Filippo Cademartiri
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| |
Collapse
|
17
|
Tian X, Fu K, Huang X, Zou H, Shi N, Li J, Bao Y, He S, Lv J. Ferroptosis in the adjuvant treatment of lung cancer-the potential of selected botanical drugs and isolated metabolites. Front Pharmacol 2024; 15:1430561. [PMID: 39193342 PMCID: PMC11347298 DOI: 10.3389/fphar.2024.1430561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Ferroptosis represents a distinct form of cell death that is not associated with necrosis, autophagy, apoptosis, or pyroptosis. It is characterised by intracellular iron-dependent lipid peroxidation. The current literature indicates that a number of botanical drugs and isolated metabolites can modulate ferroptosis, thereby exerting inhibitory effects on lung cancer cells or animal models. The aim of this review is to elucidate the mechanisms through which botanical drugs and isolated metabolites regulate ferroptosis in the context of lung cancer, thereby providing potential insights into lung cancer treatment. It is crucial to highlight that these preclinical findings should not be interpreted as evidence that these treatments can be immediately translated into clinical applications. In the future, we will continue to study the pharmacology, pharmacokinetics and toxicology of these drugs, as well as evaluating their efficacy and safety in clinical trials, with the aim of providing new approaches to the development of new agents for the treatment of lung cancer.
Collapse
Affiliation(s)
- Xiaoyan Tian
- The First Clinical Institute, Zunyi Medical University, Zunyi, Guizhou, China
| | - Kunling Fu
- The First Clinical Institute, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xuemin Huang
- The First Clinical Institute, Zunyi Medical University, Zunyi, Guizhou, China
| | - Haiyan Zou
- The First Clinical Institute, Zunyi Medical University, Zunyi, Guizhou, China
| | - Nianmei Shi
- The First Clinical Institute, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiayang Li
- Office of Drug Clinical Trial Institution, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuxiang Bao
- Department of General Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Sisi He
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Junyuan Lv
- The First Clinical Institute, Zunyi Medical University, Zunyi, Guizhou, China
- Department of General Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
18
|
Lakhal-Littleton S, Cleland JGF. Iron deficiency and supplementation in heart failure. Nat Rev Cardiol 2024; 21:463-486. [PMID: 38326440 DOI: 10.1038/s41569-024-00988-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Non-anaemic iron deficiency (NAID) is a strategic target in cardiovascular medicine because of its association with a range of adverse effects in various conditions. Endeavours to tackle NAID in heart failure have yielded mixed results, exposing knowledge gaps in how best to define 'iron deficiency' and the handling of iron therapies by the body. To address these gaps, we harness the latest understanding of the mechanisms of iron homeostasis outside the erythron and integrate clinical and preclinical lines of evidence. The emerging picture is that current definitions of iron deficiency do not assimilate the multiple influences at play in patients with heart failure and, consequently, fail to identify those with a truly unmet need for iron. Additionally, current iron supplementation therapies benefit only certain patients with heart failure, reflecting differences in the nature of the unmet need for iron and the modifying effects of anaemia and inflammation on the handling of iron therapies by the body. Building on these insights, we identify untapped opportunities in the management of NAID, including the refinement of current approaches and the development of novel strategies. Lessons learned from NAID in cardiovascular disease could ultimately translate into benefits for patients with other chronic conditions such as chronic kidney disease, chronic obstructive pulmonary disease and cancer.
Collapse
Affiliation(s)
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
19
|
Barad A, Clark AG, Pressman EK, O'Brien KO. Associations Between Genetically Predicted Iron Status and Cardiovascular Disease Risk: A Mendelian Randomization Study. J Am Heart Assoc 2024; 13:e034991. [PMID: 38818967 PMCID: PMC11255641 DOI: 10.1161/jaha.124.034991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/03/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Mendelian randomization (MR) studies suggest a causal effect of iron status on cardiovascular disease (CVD) risk, but it is unknown if these associations are confounded by pleiotropic effects of the instrumental variables on CVD risk factors. We aimed to investigate the effect of iron status on CVD risk controlling for CVD risk factors. METHODS AND RESULTS Iron biomarker instrumental variables (total iron-binding capacity [n=208 422], transferrin saturation [n=198 516], serum iron [n=236 612], ferritin [n=257 953]) were selected from a European genome-wide association study meta-analysis. We performed 2-sample univariate MR of each iron trait on CVD outcomes (all-cause ischemic stroke, cardioembolic ischemic stroke, large-artery ischemic stroke, small-vessel ischemic stroke, and coronary heart disease) from MEGASTROKE (n=440 328) and CARDIoGRAMplusC4D (Coronary Artery Disease Genome Wide Replication and Meta-Analysis Plus the Coronary Artery Disease Genetics) (n=183 305). We then implemented multivariate MR conditioning on 7 CVD risk factors from independent European samples to evaluate their potential confounding or mediating effects on the observed iron-CVD associations. With univariate MR analyses, we found higher genetically predicted iron status to be associated with a greater risk of cardioembolic ischemic stroke (transferrin saturation: odds ratio, 1.17 [95% CI, 1.03-1.33]; serum iron: odds ratio, 1.21 [95% CI, 1.02-1.44]; total iron-binding capacity: odds ratio, 0.81 [95% CI, 0.69-0.94]). The detrimental effects of iron status on cardioembolic ischemic stroke risk remained unaffected when adjusting for CVD risk factors (all P<0.05). Additionally, we found diastolic blood pressure to mediate between 7.1 and 8.8% of the total effect of iron status on cardioembolic ischemic stroke incidence. Univariate MR initially suggested a protective effect of iron status on large-artery stroke and coronary heart disease, but controlling for CVD factors using multivariate MR substantially diminished these associations (all P>0.05). CONCLUSIONS Higher iron status was associated with a greater risk of cardioembolic ischemic stroke independent of CVD risk factors, and this effect was partly mediated by diastolic blood pressure. These findings support a role of iron status as a modifiable risk factor for cardioembolic ischemic stroke.
Collapse
Affiliation(s)
- Alexa Barad
- Division of Nutritional SciencesCornell UniversityIthacaNYUSA
| | - Andrew G. Clark
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNYUSA
- Department of Computational BiologyCornell UniversityIthacaNYUSA
| | - Eva K. Pressman
- Department of Obstetrics and GynecologyUniversity of Rochester Medical CenterRochesterNYUSA
| | | |
Collapse
|
20
|
Jin S, Wang H, Zhang X, Song M, Liu B, Sun W. Emerging regulatory mechanisms in cardiovascular disease: Ferroptosis. Biomed Pharmacother 2024; 174:116457. [PMID: 38518600 DOI: 10.1016/j.biopha.2024.116457] [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: 12/21/2023] [Revised: 03/03/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
Ferroptosis, distinct from apoptosis, necrosis, autophagy, and other types of cell death, is a novel iron-dependent regulated cell death characterized by the accumulation of lipid peroxides and redox imbalance with distinct morphological, biochemical, and genetic features. Dysregulation of iron homeostasis, the disruption of antioxidative stress pathways and lipid peroxidation are crucial in ferroptosis. Ferroptosis is involved in the pathogenesis of several cardiovascular diseases, including atherosclerosis, cardiomyopathy, myocardial infarction, ischemia-reperfusion injury, abdominal aortic aneurysm, aortic dissection, and heart failure. Therefore, a comprehensive understanding of the mechanisms that regulate ferroptosis in cardiovascular diseases will enhance the prevention and treatment of these diseases. This review discusses the latest findings on the molecular mechanisms of ferroptosis and its regulation in cardiovascular diseases, the application of ferroptosis modulators in cardiovascular diseases, and the role of traditional Chinese medicines in ferroptosis regulation to provide a comprehensive understanding of the pathogenesis of cardiovascular diseases and identify new prevention and treatment options.
Collapse
Affiliation(s)
- Sijie Jin
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - Xiaohao Zhang
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - Mengyang Song
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China.
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China.
| |
Collapse
|
21
|
Zhang T, Luo L, He Q, Xiao S, Li Y, Chen J, Qin T, Xiao Z, Ge Q. Research advances on molecular mechanism and natural product therapy of iron metabolism in heart failure. Eur J Med Res 2024; 29:253. [PMID: 38659000 PMCID: PMC11044586 DOI: 10.1186/s40001-024-01809-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
The progression of heart failure (HF) is complex and involves multiple regulatory pathways. Iron ions play a crucial supportive role as a cofactor for important proteins such as hemoglobin, myoglobin, oxidative respiratory chain, and DNA synthetase, in the myocardial energy metabolism process. In recent years, numerous studies have shown that HF is associated with iron dysmetabolism, and deficiencies in iron and overload of iron can both lead to the development of various myocarditis diseases, which ultimately progress to HF. Iron toxicity and iron metabolism may be key targets for the diagnosis, treatment, and prevention of HF. Some iron chelators (such as desferrioxamine), antioxidants (such as ascorbate), Fer-1, and molecules that regulate iron levels (such as lactoferrin) have been shown to be effective in treating HF and protecting the myocardium in multiple studies. Additionally, certain natural compounds can play a significant role by mediating the imbalance of iron-related signaling pathways and expression levels. Therefore, this review not only summarizes the basic processes of iron metabolism in the body and the mechanisms by which they play a role in HF, with the aim of providing new clues and considerations for the treatment of HF, but also summarizes recent studies on natural chemical components that involve ferroptosis and its role in HF pathology, as well as the mechanisms by which naturally occurring products regulate ferroptosis in HF, with the aim of providing reference information for the development of new ferroptosis inhibitors and lead compounds for the treatment of HF in the future.
Collapse
Affiliation(s)
- Tianqing Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Li Luo
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Qi He
- People's Hospital of Ningxiang City, Ningxiang City, China
| | - Sijie Xiao
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Yuwei Li
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Junpeng Chen
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Tao Qin
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Zhenni Xiao
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Qingliang Ge
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China.
| |
Collapse
|
22
|
Fang W, Xie S, Deng W. Ferroptosis mechanisms and regulations in cardiovascular diseases in the past, present, and future. Cell Biol Toxicol 2024; 40:17. [PMID: 38509409 PMCID: PMC10955039 DOI: 10.1007/s10565-024-09853-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024]
Abstract
Cardiovascular diseases (CVDs) are the main diseases that endanger human health, and their risk factors contribute to high morbidity and a high rate of hospitalization. Cell death is the most important pathophysiology in CVDs. As one of the cell death mechanisms, ferroptosis is a new form of regulated cell death (RCD) that broadly participates in CVDs (such as myocardial infarction, heart transplantation, atherosclerosis, heart failure, ischaemia/reperfusion (I/R) injury, atrial fibrillation, cardiomyopathy (radiation-induced cardiomyopathy, diabetes cardiomyopathy, sepsis-induced cardiac injury, doxorubicin-induced cardiac injury, iron overload cardiomyopathy, and hypertrophic cardiomyopathy), and pulmonary arterial hypertension), involving in iron regulation, metabolic mechanism and lipid peroxidation. This article reviews recent research on the mechanism and regulation of ferroptosis and its relationship with the occurrence and treatment of CVDs, aiming to provide new ideas and treatment targets for the clinical diagnosis and treatment of CVDs by clarifying the latest progress in CVDs research.
Collapse
Affiliation(s)
- Wenxi Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
23
|
Barad A, Clark AG, O’Brien KO, Pressman EK. Associations between genetically predicted iron status and cardiovascular disease risk: A Mendelian randomization study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.05.24302373. [PMID: 38370765 PMCID: PMC10871385 DOI: 10.1101/2024.02.05.24302373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Background Mendelian randomization (MR) studies suggest a causal effect of iron (Fe) status on cardiovascular disease (CVD) risk, but it is unknown if these associations are confounded by pleiotropic effects of the instrumental variables (IV) on CVD risk factors. We aimed to investigate the effect of Fe status on CVD risk controlling for CVD risk factors. Methods Fe biomarker IVs (total Fe binding capacity (TIBC, n=208,422), transferrin saturation (TSAT, n=198,516), serum Fe (SI, n=236,612), ferritin (n=257,953)) were selected from a European GWAS meta-analysis. We performed two-sample univariate (UV) MR of each Fe trait on CVD outcomes (all-cause ischemic stroke (IS), cardioembolic IS (CES), large artery IS (LAS), small vessel IS (SVS), and coronary heart disease (CHD)) from MEGASTROKE (n=440,328) and CARDIoGRAMplusC4D (n=183,305). We then implemented multivariate (MV) MR conditioning on six CVD risk factors from independent European samples to evaluate their potential confounding and/or mediating effects on the observed Fe-CVD associations. Results With UVMR analyses, we found higher genetically predicted Fe status to be associated with a greater risk of CES (TSAT: OR 1.17 [95%CI 1.03, 1.33], SI: OR 1.21 [ 95%CI 1.02, 1.44]; TIBC: OR 0.81 [95%CI 0.69, 0.94]). The detrimental effects of Fe status on CES risk remained unaffected when adjusting for CVD risk factors (all P<0.05). Additionally, we found diastolic blood pressure (DBP) to mediate between 7.1-8.8% of the total effect of Fe status on CES incidence. While UVMR initially suggested a protective effect of Fe status on LAS and CHD, MVMR analyses factoring CVD risk factors revealed a complete annulment of this perceived protective effect (all P>0.05). Discussion Higher Fe status was associated with a greater risk of CES independent of CVD risk factors, and this effect was partly mediated by DBP. These findings support a role of Fe status as a modifiable risk factor for CES.
Collapse
Affiliation(s)
- Alexa Barad
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Andrew G. Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
- Department of Computational Biology, Cornell University, Ithaca, New York, USA
| | | | - Eva K. Pressman
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
24
|
Meregildo-Rodriguez ED, Asmat-Rubio MG, Bardales-Zuta VH, Vásquez-Tirado GA. Effect of calcium-channel blockers on the risk of active tuberculosis and mortality: systematic review and meta-analysis. Front Pharmacol 2024; 15:1298919. [PMID: 38303987 PMCID: PMC10830796 DOI: 10.3389/fphar.2024.1298919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024] Open
Abstract
Introduction: Recent studies suggest that calcium channel blockers (CCBs) could reduce the risk of active tuberculosis and improve clinical outcomes. We aimed to synthesize the evidence regarding the effect of CCBs on the risk of developing active tuberculosis and mortality. Methods: We systematically searched for observational studies and clinical trials published in six databases until 31 August 2023, following a PECO/PICO strategy. Results: We included eight observational studies, 4,020,830 patients, among whom 241,761 had diabetes mellitus and 30,397 had active tuberculosis. According to our results, CCBs reduce the risk of developing active tuberculosis by 29% (RR 0.71; 95% CI 0.67-0.75) in patients with and without diabetes mellitus. However, CCBs do not show any benefit in terms of tuberculosis-related mortality (RR 1.00; 95% CI 0.98-1.02). For both outcomes, no statistical heterogeneity was found (I2 = 0, p > 0.10). This protective effect of CCBs on the risk of active tuberculosis remained independent of the type of patient (with diabetes mellitus vs. general population) or the class of CCB administered (DHP-CCB vs. non-DHP-CCB) (test for subgroup differences I2 = 0, p > 0.10). However, this beneficial effect was more significant among the general population (RR 0.70; 95% CI 0.66-0.74) compared to patients with diabetes mellitus (RR 0.72; 95% CI 0.61-0.86) and among those patients treated with DHP-CCBs (RR 0.69; 95% CI 0.63-0.74) compared to patients treated with non-DHP-CCBs (RR 0.72; 95% CI 0.67-0.78). Conclusion: CCBs may reduce the risk of active TB in patients with diabetes and the general population. On the contrary, CCBs do not seem to have a protective effect on tuberculosis-related mortality. However, more evidence is still needed. We recommend developing clinical trials to verify these findings, including more diverse populations. Systematic Review Registration: [https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=352129].
Collapse
|
25
|
Liu G, Xie X, Liao W, Chen S, Zhong R, Qin J, He P, Xie J. Ferroptosis in cardiovascular disease. Biomed Pharmacother 2024; 170:116057. [PMID: 38159373 DOI: 10.1016/j.biopha.2023.116057] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
In the 21st century, cardiovascular disease (CVD) has become one of the leading causes of death worldwide. The prevention and treatment of CVD remain pressing scientific issues. Several recent studies have suggested that ferroptosis may play a key role in CVD. Most studies conducted thus far on ferroptosis and CVD have supported the link. Ferroptosis mediated by different signaling and metabolic pathways can lead to ischemic heart disease, myocarditis, heart failure, ischemia-reperfusion injury, and cardiomyopathy. Still, the specific mechanism of ferroptosis in CVD, the particular organ areas affected, and the stage of disease involved need to be further studied. Therefore, understanding the mechanisms regulating ferroptosis in CVD may improve disease management. Throughout this review, we summarized the mechanism of ferroptosis and its effect on the pathogenesis of CVD. We also predicted and discussed future research directions, aiming to provide new ideas and strategies for preventing and treating CVD.
Collapse
Affiliation(s)
- Guoqing Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoyong Xie
- Departments of Pathophysiology, Guangxi Medical University, Nanning, Guangxi, China
| | - Wang Liao
- Department of Cardiology, The First People's Hospital of Yulin, Yulin, Guangxi, China
| | - Siyuan Chen
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Rumao Zhong
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiahui Qin
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Peichun He
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jian Xie
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
26
|
Meloni A, Pistoia L, Vassalle C, Spasiano A, Fotzi I, Bagnato S, Putti MC, Cossu A, Massei F, Giovangrossi P, Maffei S, Positano V, Cademartiri F. Low Vitamin D Levels Are Associated with Increased Cardiac Iron Uptake in Beta-Thalassemia Major. Diagnostics (Basel) 2023; 13:3656. [PMID: 38132240 PMCID: PMC10742632 DOI: 10.3390/diagnostics13243656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
We evaluated the association of vitamin D and parathormone (PTH) levels with cardiac iron and function in beta-thalassemia major (β-TM) patients. Two-hundred and seventy-eight TM patients (39.04 ± 8.58 years, 56.8% females) underwent magnetic resonance imaging for the assessment of iron overload (T2* technique), biventricular function parameters (cine images), and replacement myocardial fibrosis (late gadolinium enhancement technique). Vitamin D levels were deficient (<20 ng/dL) in 107 (38.5%) patients, insufficient (20-30 ng/dL) in 96 (34.5%) patients, and sufficient (≥30 ng/dL) in 75 (27.0%) patients. Deficient vitamin D patients had a significantly higher frequency of myocardial iron overload (MIO; global heart T2* < 20 ms) than patients with sufficient and insufficient vitamin D levels and a significantly higher left ventricular end-diastolic volume index and mass index than patients with sufficient vitamin D levels. PTH was not associated with cardiac iron, function, or fibrosis. In the multivariate regression analysis, vitamin D, serum ferritin, and pancreatic iron levels were the strongest predictors of global heart T2* values. In receiver operating characteristic curve analysis, a vitamin D level ≤ 17.3 ng/dL predicted MIO with a sensitivity of 81.5% and a specificity of 75.3% (p < 0.0001). In TM, the periodic and regular assessment of vitamin D levels can be beneficial for the prevention of cardiac iron accumulation and subsequent overt dysfunction.
Collapse
Affiliation(s)
- Antonella Meloni
- Bioengineering Unit, Fondazione G. Monasterio CNR—Regione Toscana, 56124 Pisa, Italy; (A.M.); (V.P.)
- Department of Radiology, Fondazione G. Monasterio CNR—Regione Toscana, 56124 Pisa, Italy;
| | - Laura Pistoia
- Department of Radiology, Fondazione G. Monasterio CNR—Regione Toscana, 56124 Pisa, Italy;
- Unità Operativa Complessa Ricerca Clinica, Fondazione G. Monasterio CNR—Regione Toscana, 56124 Pisa, Italy
| | - Cristina Vassalle
- Medicina di Laboratorio, Fondazione G. Monasterio CNR—Regione Toscana, 56124 Pisa, Italy;
| | - Anna Spasiano
- Unità Operativa Semplice Dipartimentale Malattie Rare del Globulo Rosso, Azienda Ospedaliera di Rilievo Nazionale “A. Cardarelli”, 80131 Napoli, Italy;
| | - Ilaria Fotzi
- Oncologia, Ematologia e Trapianto di Cellule Staminali Emopoietiche, Meyer Children’s Hospital IRCCS, 50139 Firenze, Italy;
| | - Sergio Bagnato
- Ematologia Microcitemia, Ospedale San Giovanni di Dio—ASP Crotone, 88900 Crotone, Italy;
| | - Maria Caterina Putti
- Dipartimento della Salute della Donna e del Bambino, Clinica di Emato-Oncologia Pediatrica, Azienda Ospedaliero Università di Padova, 35128 Padova, Italy;
| | - Antonella Cossu
- Ambulatorio Trasfusionale—Servizio Immunoematologia e Medicina Trasfusionale Dipartimento dei Servizi, Presidio Ospedaliero “San Francesco”, 08100 Nuoro, Italy;
| | - Francesco Massei
- Unità Operativa Oncoematologia Pediatrica, Azienda Ospedaliero Universitaria Pisana—Stabilimento S. Chiara, 56126 Pisa, Italy;
| | - Piera Giovangrossi
- Servizio di Immunoematologia e Medicina Trasfusionale, Ospedale S. M. Goretti, 04100 Latina, Italy;
| | - Silvia Maffei
- Cardiovascular and Gynaecological Endocrinology Unit, Fondazione G. Monasterio CNR—Regione Toscana, 56124 Pisa, Italy;
| | - Vincenzo Positano
- Bioengineering Unit, Fondazione G. Monasterio CNR—Regione Toscana, 56124 Pisa, Italy; (A.M.); (V.P.)
- Department of Radiology, Fondazione G. Monasterio CNR—Regione Toscana, 56124 Pisa, Italy;
| | - Filippo Cademartiri
- Department of Radiology, Fondazione G. Monasterio CNR—Regione Toscana, 56124 Pisa, Italy;
| |
Collapse
|
27
|
Gawargi FI, Mishra PK. Ironing out the details: ferroptosis and its relevance to diabetic cardiomyopathy. Am J Physiol Regul Integr Comp Physiol 2023; 325:R665-R681. [PMID: 37746707 PMCID: PMC11178299 DOI: 10.1152/ajpregu.00117.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
Ferroptosis is a newly identified myocardial cell death mechanism driven by iron-dependent lipid peroxidation. The presence of elevated intramyocardial lipid levels and excessive iron in patients with diabetes suggest a predominant role of ferroptosis in diabetic cardiomyopathy. As myocardial cell death is a precursor of heart failure, and intensive glycemic control cannot abate the increased risk of heart failure in patients with diabetes, targeting myocardial cell death via ferroptosis is a promising therapeutic avenue to prevent and/or treat diabetic cardiomyopathy. This review provides updated and comprehensive molecular mechanisms underpinning ferroptosis, clarifies several misconceptions about ferroptosis, emphasizes the importance of ferroptosis in diabetes-induced myocardial cell death, and offers valuable approaches to evaluate and target ferroptosis in the diabetic heart. Furthermore, basic concepts and ideas presented in this review, including glutathione peroxidase-4-independent and mitochondrial mechanisms of ferroptosis, are also important for investigating ferroptosis in other diabetic organs, as well as nondiabetic and metabolically compromised hearts.
Collapse
Affiliation(s)
- Flobater I Gawargi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| |
Collapse
|
28
|
Padhani ZA, Gangwani MK, Sadaf A, Hasan B, Colan S, Alvi N, Das JK. Calcium channel blockers for preventing cardiomyopathy due to iron overload in people with transfusion-dependent beta thalassaemia. Cochrane Database Syst Rev 2023; 11:CD011626. [PMID: 37975597 PMCID: PMC10655499 DOI: 10.1002/14651858.cd011626.pub3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
BACKGROUND Beta-thalassaemia is an inherited blood disorder that reduces the production of haemoglobin. The most severe form requires recurrent blood transfusions, which can lead to iron overload. Cardiovascular dysfunction caused by iron overload is the leading cause of morbidity and mortality in people with transfusion-dependent beta-thalassaemia. Iron chelation therapy has reduced the severity of systemic iron overload, but removal of iron from the myocardium requires a very proactive preventive strategy. There is evidence that calcium channel blockers may reduce myocardial iron deposition. This is an update of a Cochrane Review first published in 2018. OBJECTIVES To assess the effects of calcium channel blockers plus standard iron chelation therapy, compared with standard iron chelation therapy (alone or with a placebo), on cardiomyopathy due to iron overload in people with transfusion-dependent beta thalassaemia. SEARCH METHODS We searched the Cochrane Haemoglobinopathies Trials Register, compiled from electronic database searches and handsearching of journals and conference abstract books, to 13 January 2022. We also searched ongoing trials databases and the reference lists of relevant articles and reviews. SELECTION CRITERIA We included randomised controlled trials (RCTs) of calcium channel blockers combined with standard chelation therapy versus standard chelation therapy alone or combined with placebo in people with transfusion-dependent beta thalassaemia. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. We used GRADE to assess certainty of evidence. MAIN RESULTS We included six RCTs (five parallel-group trials and one cross-over trial) with 253 participants; there were 126 participants in the amlodipine arms and 127 in the control arms. The certainty of the evidence was low for most outcomes at 12 months; the evidence for liver iron concentration was of moderate certainty, and the evidence for adverse events was of very low certainty. Amlodipine plus standard iron chelation compared with standard iron chelation (alone or with placebo) may have little or no effect on cardiac T2* values at 12 months (mean difference (MD) 1.30 ms, 95% confidence interval (CI) -0.53 to 3.14; 4 trials, 191 participants; low-certainty evidence) and left ventricular ejection fraction (LVEF) at 12 months (MD 0.81%, 95% CI -0.92% to 2.54%; 3 trials, 136 participants; low-certainty evidence). Amlodipine plus standard iron chelation compared with standard iron chelation (alone or with placebo) may reduce myocardial iron concentration (MIC) after 12 months (MD -0.27 mg/g, 95% CI -0.46 to -0.08; 3 trials, 138 participants; low-certainty evidence). The results of our analysis suggest that amlodipine has little or no effect on heart T2*, MIC, or LVEF after six months, but the evidence is very uncertain. Amlodipine plus standard iron chelation compared with standard iron chelation (alone or with placebo) may increase liver T2* values after 12 months (MD 1.48 ms, 95% CI 0.27 to 2.69; 3 trials, 127 participants; low-certainty evidence), but may have little or no effect on serum ferritin at 12 months (MD 0.07 μg/mL, 95% CI -0.20 to 0.35; 4 trials, 187 participants; low-certainty evidence), and probably has little or no effect on liver iron concentration (LIC) after 12 months (MD -0.86 mg/g, 95% CI -4.39 to 2.66; 2 trials, 123 participants; moderate-certainty evidence). The results of our analysis suggest that amlodipine has little or no effect on serum ferritin, liver T2* values, or LIC after six months, but the evidence is very uncertain. The included trials did not report any serious adverse events at six or 12 months of intervention. The studies did report mild adverse effects such as oedema, dizziness, mild cutaneous allergy, joint swelling, and mild gastrointestinal symptoms. Amlodipine may be associated with a higher risk of oedema (risk ratio (RR) 5.54, 95% CI 1.24 to 24.76; 4 trials, 167 participants; very low-certainty evidence). We found no difference between the groups in the occurrence of other adverse events, but the evidence was very uncertain. No trials reported mortality, cardiac function assessments other than echocardiographic estimation of LVEF, electrocardiographic abnormalities, quality of life, compliance with treatment, or cost of interventions. AUTHORS' CONCLUSIONS The available evidence suggests that calcium channel blockers may reduce MIC and may increase liver T2* values in people with transfusion-dependent beta thalassaemia. Longer-term multicentre RCTs are needed to assess the efficacy and safety of calcium channel blockers for myocardial iron overload, especially in younger children. Future trials should also investigate the role of baseline MIC in the response to calcium channel blockers, and include a cost-effectiveness analysis.
Collapse
Affiliation(s)
- Zahra Ali Padhani
- Institute for Global Health and Development, Aga Khan University, Karachi, Pakistan
- Robinson Research Institute, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Karachi, Pakistan
| | | | - Alina Sadaf
- Department of Paediatric Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, Pakistan
| | - Babar Hasan
- Division of Cardiothoracic Sciences, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Steven Colan
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Najveen Alvi
- Department of Pediatrics, Aga Khan University, Karachi, Pakistan
| | - Jai K Das
- Institute for Global Health and Development, Aga Khan University Hospital, Karachi, Pakistan
- Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
29
|
Kumfu S, Sripetchwandee J, Thonusin C, Sumneang N, Maneechote C, Arunsak B, Chunchai T, Oo TT, Kongkaew A, Chattipakorn SC, Chattipakorn N. Ferroptosis inhibitor improves cardiac function more effectively than inhibitors of apoptosis and necroptosis through cardiac mitochondrial protection in rats with iron-overloaded cardiomyopathy. Toxicol Appl Pharmacol 2023; 479:116727. [PMID: 37863361 DOI: 10.1016/j.taap.2023.116727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Iron overload cardiomyopathy (IOC) is the leading cause of death in cases of iron overload in patients. Previous studies demonstrated that iron overload led to cardiomyocyte dysfunction and death through multiple pathways including apoptosis, necroptosis and ferroptosis. However, the dominant cell death pathway in the iron-overloaded heart needs clarification. We tested the hypothesis that ferroptosis, an iron-dependent cell death, plays a dominant role in IOC, and ferroptosis inhibitor exerts greater efficacy than inhibitors of apoptosis and necroptosis on improving cardiac function in iron-overloaded rats. Iron dextran was injected intraperitoneally into male Wistar rats for four weeks to induce iron overload. Then, the rats were divided into 5 groups: treated with vehicle, apoptosis inhibitor (z-VAD-FMK), necroptosis inhibitor (Necrostatin-1), ferroptosis inhibitor (Ferrostatin-1) or iron chelator (deferoxamine) for 2 weeks. Cardiac function, mitochondrial function, apoptosis, necroptosis and ferroptosis were determined. The increased expression of apoptosis-, necroptosis- and ferroptosis-related proteins, were associated with impaired cardiac and mitochondrial function in iron-overloaded rats. All cell death inhibitors attenuated cardiac apoptosis, necroptosis and ferroptosis in iron-overloaded rats. Ferrostatin-1 was more effective than the other drugs in diminishing mitochondrial dysfunction and Bax/Bcl-2 ratio. Moreover, both Ferrostatin-1 and deferoxamine reversed iron overload-induced cardiac dysfunction as indicated by restored left ventricular ejection fraction and E/A ratio, whereas z-VAD-FMK and Necrostatin-1 only partially improved this parameter. These results indicated that ferroptosis could be the predominant form of cardiomyocyte death in IOC, and that inhibiting ferroptosis might be a potential novel treatment for IOC.
Collapse
Affiliation(s)
- Sirinart Kumfu
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Jirapas Sripetchwandee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Chanisa Thonusin
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Natticha Sumneang
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Thura Tun Oo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
30
|
Khuanjing T, Maneechote C, Ongnok B, Prathumsap N, Arinno A, Chunchai T, Arunsak B, Chattipakorn SC, Chattipakorn N. Vagus nerve stimulation and acetylcholinesterase inhibitor donepezil provide cardioprotection against trastuzumab-induced cardiotoxicity in rats by attenuating mitochondrial dysfunction. Biochem Pharmacol 2023; 217:115836. [PMID: 37816466 DOI: 10.1016/j.bcp.2023.115836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/24/2023] [Accepted: 09/27/2023] [Indexed: 10/12/2023]
Abstract
Trastuzumab (Trz) is a targeted anticancer drug for human epidermal growth factor receptor 2 (HER2)-positive tumors, as Trz-induced cardiotoxicity (TIC) is commonly observed in Trz-treated patients. Since cardiac autonomic modulation with electrical vagus nerve stimulation (VNS) and acetylcholinesterase (AChE) inhibitors exerts cardioprotection against various heart diseases, the comparative effects of electrical VNS and an AChE inhibitor (donepezil) on cardiac and mitochondrial functions and programmed cell death pathways in TIC are not known. VNS devices were implanted in thirty-two male Wistar rats and were divided into 4 groups: (i) Control-Sham (CSham), (ii) Trz-Sham (TSham), (iii) Trz-VNS (TVNS), and (iv) Trz-donepezil (TDPZ). Rats in the Trz-treated groups were intraperitoneally injected with Trz (4 mg/kg/day) for 7 days, while CSham rats were injected with NSS. VNS devices were activated in the TVNS rats during the 7-day Trz treatment, but not in the sham rats. Rats in the TDPZ group received donepezil orally (5 mg/kg/day) for 7 days. At the end, left ventricular (LV) function and heart rate variability were evaluated, and heart tissue was collected for biochemical and histological analysis. Trz rats showed LV dysfunction and cardiac sympathovagal imbalance. In addition, mitochondrial function and dynamics were impaired in TIC rats. Trz also increased cardiomyocyte death by inducing apoptosis, pyroptosis, and ferroptosis. Electrical VNS and donepezil had similar efficacy in alleviating cardiac mitochondrial dysfunction, dynamic imbalances, and cardiomyocyte death, leading to improved LV function. These findings suggested that parasympathetic activation via either VNS or an AChE inhibitor could be a promising therapeutic intervention against TIC.
Collapse
Affiliation(s)
- Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Benjamin Ongnok
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nanthip Prathumsap
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Apiwan Arinno
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
31
|
Zhao K, Chen X, Bian Y, Zhou Z, Wei X, Zhang J. Broadening horizons: The role of ferroptosis in myocardial ischemia-reperfusion injury. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2269-2286. [PMID: 37119287 DOI: 10.1007/s00210-023-02506-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/19/2023] [Indexed: 05/01/2023]
Abstract
Ferroptosis is a novel type of regulated cell death (RCD) discovered in recent years, where abnormal intracellular iron accumulation leads to the onset of lipid peroxidation, which further leads to the disruption of intracellular redox homeostasis and triggers cell death. Iron accumulation with lipid peroxidation is considered a hallmark of ferroptosis that distinguishes it from other RCDs. Myocardial ischemia-reperfusion injury (MIRI) is a process of increased myocardial cell injury that occurs during coronary reperfusion after myocardial ischemia and is associated with high post-infarction mortality. Multiple experiments have shown that ferroptosis plays an important role in MIRI pathophysiology. This review systematically summarized the latest research progress on the mechanisms of ferroptosis. Then we report the possible link between the occurrence of MIRI and ferroptosis in cardiomyocytes. Finally, we discuss and analyze the related drugs that target ferroptosis to attenuate MIRI and its action targets, and point out the shortcomings of the current state of relevant research and possible future research directions. It is hoped to provide a new avenue for improving the prognosis of the acute coronary syndrome.
Collapse
Affiliation(s)
- Ke Zhao
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Xiaoshu Chen
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yujing Bian
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Zhou Zhou
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Xijin Wei
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, China.
| | - Juan Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, China.
| |
Collapse
|
32
|
Meloni A, Pistoia L, Ricchi P, Positano V, Longo F, Borsellino Z, Cecinati V, Messina G, Corigliano E, Rosso R, Righi R, Peritore G, Renne S, Vallone A, Cademartiri F. Pancreatic T2* Magnetic Resonance Imaging for Prediction of Cardiac Arrhythmias in Transfusion-Dependent Thalassemia. J Clin Med 2023; 12:6015. [PMID: 37762955 PMCID: PMC10531669 DOI: 10.3390/jcm12186015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
We assessed the value of pancreatic T2* magnetic resonance imaging (MRI) for predicting cardiac events from a large prospective database of transfusion-dependent thalassemia (TDT) patients. We considered 813 TDT patients (36.47 ± 10.71 years, 54.6% females) enrolled in the Extension-Myocardial Iron Overload in Thalassemia Network. MRI was used to measure hepatic, pancreatic, and cardiac iron overload (IO), to assess biventricular function and atrial dimensions, and to detect replacement myocardial fibrosis. The mean follow-up was 50.51 ± 19.75 months. Cardiac complications were recorded in 21 (2.6%) patients: one with heart failure (HF) and 20 with arrhythmias. The single patient who developed HF had, at the baseline MRI, a reduced pancreas T2*. Out of the 20 recorded arrhythmias, 17 were supraventricular. Pancreatic T2* values were a significant predictor of future arrhythmia-related events (hazard ratio = 0.89; p = 0.015). Pancreas T2* remained significantly associated with future arrhythmias after adjusting for any other univariate predictor (age and male sex, diabetes, history of previous arrhythmias, or left atrial area index). According to the receiver-operating characteristic curve analysis for arrhythmias, a pancreas T2* < 6.73 ms was the optimal cut-off value. In TDT, pancreatic iron levels had significant prognostic power for arrhythmias. Regular monitoring and the development of targeted interventions to manage pancreatic IO may help improve patient outcomes.
Collapse
Affiliation(s)
- Antonella Meloni
- Department of Radiology, Fondazione Gabriele Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.)
- Unità Operativa Complessa Bioingegneria, Fondazione Gabriele Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | - Laura Pistoia
- Department of Radiology, Fondazione Gabriele Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.)
- Unità Operativa Semplice a Valenza Dipartimentale Ricerca Clinica, Fondazione Gabriele Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | - Paolo Ricchi
- Unità Operativa Semplice Dipartimentale Malattie Rare del Globulo Rosso, Azienda Ospedaliera di Rilievo Nazionale “Antonio Cardarelli”, 80131 Napoli, Italy;
| | - Vincenzo Positano
- Department of Radiology, Fondazione Gabriele Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.)
- Unità Operativa Complessa Bioingegneria, Fondazione Gabriele Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | - Filomena Longo
- Unità Operativa Day Hospital della Talassemia e delle Emoglobinopatie, Azienda Ospedaliero-Universitaria “S. Anna”, 44124 Cona, Italy;
| | - Zelia Borsellino
- Unità Operativa Complessa Ematologia con Talassemia, ARNAS Civico “Benfratelli-Di Cristina”, 90134 Palermo, Italy;
| | - Valerio Cecinati
- Struttura Semplice di Microcitemia, Ospedale “Santissima Annunziata”, 74123 Taranto, Italy;
| | - Giuseppe Messina
- Centro Microcitemie, Grande Ospedale Metropolitano “Bianchi-Melacrino-Morelli”, 89100 Reggio Calabria, Italy;
| | - Elisabetta Corigliano
- Ematologia Microcitemia, Ospedale San Giovanni di Dio—ASP Crotone, 88900 Crotone, Italy;
| | - Rosamaria Rosso
- Unità Operativa Talassemie ed Emoglobinopatie, Azienda Ospedaliero-Universitaria Policlinico “Vittorio Emanuele”, 95100 Catania, Italy;
| | - Riccardo Righi
- Diagnostica per Immagini e Radiologia Interventistica, Ospedale del Delta, 44023 Lagosanto, Italy;
| | - Giuseppe Peritore
- Unità Operativa Complessa di Radiologia, ARNAS Civico “Benfratelli-Di Cristina”, 90127 Palermo, Italy;
| | - Stefania Renne
- Struttura Complessa di Cardioradiologia-UTIC, Presidio Ospedaliero “Giovanni Paolo II”, 88046 Lamezia Terme, Italy;
| | - Antonino Vallone
- Reparto di Radiologia, Azienda Ospedaliera “Garibaldi” Presidio Ospedaliero Nesima, 95126 Catania, Italy;
| | - Filippo Cademartiri
- Department of Radiology, Fondazione Gabriele Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.)
| |
Collapse
|
33
|
Sheng SY, Li JM, Hu XY, Wang Y. Regulated cell death pathways in cardiomyopathy. Acta Pharmacol Sin 2023; 44:1521-1535. [PMID: 36914852 PMCID: PMC10374591 DOI: 10.1038/s41401-023-01068-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
Heart disease is a worldwide health menace. Both intractable primary and secondary cardiomyopathies contribute to malignant cardiac dysfunction and mortality. One of the key cellular processes associated with cardiomyopathy is cardiomyocyte death. Cardiomyocytes are terminally differentiated cells with very limited regenerative capacity. Various insults can lead to irreversible damage of cardiomyocytes, contributing to progression of cardiac dysfunction. Accumulating evidence indicates that majority of cardiomyocyte death is executed by regulating molecular pathways, including apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Importantly, these forms of regulated cell death (RCD) are cardinal features in the pathogenesis of various cardiomyopathies, including dilated cardiomyopathy, diabetic cardiomyopathy, sepsis-induced cardiomyopathy, and drug-induced cardiomyopathy. The relevance between abnormity of RCD with adverse outcome of cardiomyopathy has been unequivocally evident. Therefore, there is an urgent need to uncover the molecular and cellular mechanisms for RCD in order to better understand the pathogenesis of cardiomyopathies. In this review, we summarize the latest progress from studies on RCD pathways in cardiomyocytes in context of the pathogenesis of cardiomyopathies, with particular emphasis on apoptosis, necroptosis, ferroptosis, autophagy, and pyroptosis. We also elaborate the crosstalk among various forms of RCD in pathologically stressed myocardium and the prospects of therapeutic applications targeted to various cell death pathways.
Collapse
Affiliation(s)
- Shu-Yuan Sheng
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Jia-Min Li
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Xin-Yang Hu
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Yibin Wang
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China.
- Signature Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School and National Heart Center of Singapore, Singapore, Singapore.
| |
Collapse
|
34
|
Sayani FA, Singer ST, Carlberg KT, Vichinsky EP. Fertility and Pregnancy in Women with Transfusion-Dependent Thalassemia. Hematol Oncol Clin North Am 2023; 37:393-411. [PMID: 36907611 DOI: 10.1016/j.hoc.2022.12.008] [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: 03/12/2023]
Abstract
Because women with transfusion-dependent thalassemia are seeking pregnancy, ensuring the best outcomes for both mother and baby require concerted and collaborative efforts between the hematologist, obstetrician, cardiologist, hepatologist, and genetic counselor among others. Proactive counseling, early fertility evaluation, optimal management of iron overload and organ function, and application of advances in reproductive technology and prenatal screening are important in ensuring a healthy outcome. Many unanswered questions remain requiring further study, including fertility preservation, non-invasive prenatal diagnosis, chelation therapy during pregnancy, and indications and duration of anticoagulation.
Collapse
Affiliation(s)
- Farzana A Sayani
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania, Perelman School of Medicine, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Sylvia T Singer
- Division of Hematology/Oncology, UCSF Benioff Children's Hospital, 747 52nd Street, Oakland, CA 94609, USA
| | - Katie T Carlberg
- Division of Cancer and Blood Disorders, University of Washington, Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA 98105, USA
| | - Elliott P Vichinsky
- Division of Hematology/Oncology, UCSF Benioff Children's Hospital, 747 52nd Street, Oakland, CA 94609, USA
| |
Collapse
|
35
|
Soliman Y, Abdelaziz A, Mouffokes A, Amer BE, Goudy YM, Abdelwahab OA, Badawy MM, Diab RA, Elsharkawy A. Efficacy and safety of calcium channel blockers in preventing cardiac siderosis in thalassemia patients: An updated meta-analysis with trial sequential analysis. Eur J Haematol 2023; 110:414-425. [PMID: 36565288 DOI: 10.1111/ejh.13919] [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: 10/01/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Iron overload in patients with thalassemia represents a serious complication by affecting numerous organ systems. This meta-analysis aims to establish an evidence regarding the effect of amlodipine on cardiac iron overload in thalassemia patients. METHODS We searched PubMed, Scopus, Web of Science, Cochrane Central, and EMBASE for all relevant randomized controlled trials (RCTs). The primary outcomes were cardiac T2* and myocardial iron concentration (MIC). Secondary outcomes were liver iron concentration (LIC), risk of Gastrointestinal (G.I.) upset and risk of lower limb edema. We used Hedges' g to pool continuous outcomes, while odds ratio was used for dichotomous outcomes. RESULTS Seven RCTs were eligible for this systematic review and meta-analysis, comprising of 233 patients included in the analysis. Amlodipine had a statistically significant lower MIC (Hedges' g = -0.82, 95% confidence interval [CI] [-1.40, -0.24], p < .001) and higher cardiac T2* (Hedges' g = 0.36, 95% CI [0.10, 0.62], p = .03). Amlodipine was comparable to standard chelation therapy in terms of the risk of lower limb edema and GI upset. CONCLUSION Our meta-analysis found that amlodipine significantly increases cardiac T2* and decreases MIC, hence decreasing the incidence of cardiomyopathy-related iron overload in thalassemia patients.
Collapse
Affiliation(s)
- Youssef Soliman
- Medical Research Group of Egypt, Egypt.,Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed Abdelaziz
- Medical Research Group of Egypt, Egypt.,Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Adel Mouffokes
- Medical Research Group of Egypt, Egypt.,Faculty of Medicine, University of Oran 1 Ahmed Ben Bella, Oran, Algeria
| | - Basma E Amer
- Medical Research Group of Egypt, Egypt.,Faculty of Medicine, Benha University, Banha, Egypt
| | - Yomna Mohamed Goudy
- Medical Research Group of Egypt, Egypt.,Faculty of Medicine, South Valley University, Qena, Egypt
| | - Omar Ahmed Abdelwahab
- Medical Research Group of Egypt, Egypt.,Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Marwa M Badawy
- Medical Research Group of Egypt, Egypt.,Faculty of Medicine, October 6 University, Giza, Egypt
| | - Rehab Adel Diab
- Medical Research Group of Egypt, Egypt.,Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Asmaa Elsharkawy
- Medical Research Group of Egypt, Egypt.,Department of Pediatrics, Hematology and Oncology Unit, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
36
|
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: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [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
|
37
|
Sun 孙意冉 Y, Yan C, He L, Xiang S, Wang P, Li Z, Chen Y, Zhao J, Yuan Y, Wang W, Zhang X, Su P, Su Y, Ma J, Xu J, Peng Q, Ma H, Xie Z, Zhang Z. Inhibition of ferroptosis through regulating neuronal calcium homeostasis: An emerging therapeutic target for Alzheimer's disease. Ageing Res Rev 2023; 87:101899. [PMID: 36871781 DOI: 10.1016/j.arr.2023.101899] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Alzheimer's disease (AD), a chronic and progressive neurodegenerative disease, generates a serious threat to the health of the elderly. The AD brain is microscopically characterized by amyloid plaques and neurofibrillary tangles. There are still no effective therapeutic drugs to restrain the progression of AD though much attention has been paid to exploit AD treatments. Ferroptosis, a type of programmed cell death, has been reported to promote the pathological occurrence and development of AD, and inhibition of neuronal ferroptosis can effectively improve the cognitive impairment of AD. Studies have shown that calcium (Ca2+) dyshomeostasis is closely related to the pathology of AD, and can drive the occurrence of ferroptosis through several pathways, such as interacting with iron, and regulating the crosstalk between endoplasmic reticulum (ER) and mitochondria. This paper mainly reviews the roles of ferroptosis and Ca2+ in the pathology of AD, and highlights that restraining ferroptosis through maintaining the homeostasis of Ca2+ may be an innovative target for the treatment of AD.
Collapse
Affiliation(s)
- Yiran Sun 孙意冉
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Chenchen Yan
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Libo He
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China
| | - Shixie Xiang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Pan Wang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Zhonghua Li
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yuanzhao Chen
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jie Zhao
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ye Yuan
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Wang Wang
- School of basic medicine, Nanchang Medical College, Nanchang 330052, Jiangxi, China
| | - Xiaowei Zhang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Pan Su
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yunfang Su
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jinlian Ma
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jiangyan Xu
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Quekun Peng
- School of Biosciences and Technology, Chengdu Medical College, Chengdu 610500, China.
| | - Huifen Ma
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhishen Xie
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhenqiang Zhang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
38
|
Ju H, Liu T, Yang M, Cheng M, Wu G. Iron and atrial fibrillation: A review. Pacing Clin Electrophysiol 2023; 46:312-318. [PMID: 36799332 DOI: 10.1111/pace.14678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
Atrial fibrillation (AF), one of the most common arrhythmias in clinical practice, is classified into paroxysmal, persistent, and permanent AF according to its duration. The development of AF is associated with increased cardiovascular morbidity and mortality. However, the exact etiology of this disease remains poorly understood. Recent studies found disorders of iron metabolism might be involved in the progression of AF. Abnormal iron metabolism in cardiomyocytes provides arrhythmogenic substrates through a variety of mechanisms, including calcium mishandling, ion channel remodeling, and oxidative stress overaction. Interestingly, in AF patients with iron overload, interventions on iron metabolism, such as iron chelators and ferroptosis inhibitors, has been shown to prevent AF via reducing ferroptosis. Herein, we review the possible mechanisms, consequences, and therapeutic implications of altered atrial iron handling for AF pathophysiology.
Collapse
Affiliation(s)
- Hao Ju
- Department of Cardiology, Remin Hospital of Wuhan University, Wuhan, China
| | - Tao Liu
- Department of Cardiology, Remin Hospital of Wuhan University, Wuhan, China
| | - Manqi Yang
- Department of Cardiology, Remin Hospital of Wuhan University, Wuhan, China
| | - Mian Cheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Gang Wu
- Department of Cardiology, Remin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
39
|
Abstract
The cardiovascular system requires iron to maintain its high energy demands and metabolic activity. Iron plays a critical role in oxygen transport and storage, mitochondrial function, and enzyme activity. However, excess iron is also cardiotoxic due to its ability to catalyze the formation of reactive oxygen species and promote oxidative damage. While mammalian cells have several redundant iron import mechanisms, they are equipped with a single iron-exporting protein, which makes the cardiovascular system particularly sensitive to iron overload. As a result, iron levels are tightly regulated at many levels to maintain homeostasis. Iron dysregulation ranges from iron deficiency to iron overload and is seen in many types of cardiovascular disease, including heart failure, myocardial infarction, anthracycline-induced cardiotoxicity, and Friedreich's ataxia. Recently, the use of intravenous iron therapy has been advocated in patients with heart failure and certain criteria for iron deficiency. Here, we provide an overview of systemic and cellular iron homeostasis in the context of cardiovascular physiology, iron deficiency, and iron overload in cardiovascular disease, current therapeutic strategies, and future perspectives.
Collapse
Affiliation(s)
- Konrad Teodor Sawicki
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Adam De Jesus
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
| | - Hossein Ardehali
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
40
|
Zhabyeyev P, Sadasivan C, Shah S, Wang F, Oudit GY. Amlodipine rescues advanced iron overload cardiomyopathy in hemojuvelin knockout murine model: Clinical implications. Front Cardiovasc Med 2023; 10:1129349. [PMID: 37153462 PMCID: PMC10160373 DOI: 10.3389/fcvm.2023.1129349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/28/2023] [Indexed: 05/09/2023] Open
Abstract
Background Iron overload cardiomyopathy (IOC) is a major co-morbidity of genetic hemochromatosis and secondary iron overload with limited therapeutic options. We aim to investigate mechanisms of rescue action of amlodipine in the murine model of iron overload, characterize changes in human cardiac tissue due to IOC, and compare them to the changes in the animal model of IOC. Methods and results As an animal model, we used male hemojuvelin knockout (HJVKO) mice, which lacked hemojuvelin (a co-receptor protein for hepcidin expression). The mice were fed a high-iron diet from 4 weeks to 1 year of age. As a rescue, iron-fed mice received the Ca2+ channel blocker, amlodipine, from 9 to 12 months. Iron overload resulted in systolic and diastolic dysfunctions and changes in the cardiac tissue similar to the changes in the explanted human heart with IOC. An IOC patient (β-thalassemia) with left-ventricular ejection fraction (LVEF) 25% underwent heart transplantation. The murine model and the explanted heart showed intra-myocyte iron deposition, fibrosis, hypertrophy, oxidative stress, remodeling of Ca2+ cycling proteins, and metabolic kinases typical of heart failure. Single-myocyte contractility and Ca2+ release were diminished in the murine model. The amlodipine-treated group exhibited normalization of cellular function and reversed fibrosis, hypertrophy, oxidative stress, and metabolic remodeling. We also report a clinical case of primary hemochromatosis successfully treated with amlodipine. Conclusions The aged HJVKO murine model on the iron-rich diet reproduced many features of the human case of IOC. The use of amlodipine in the murine model and clinical case reversed IOC remodeling, demonstrating that amlodipine is effective adjuvant therapy for IOC.
Collapse
Affiliation(s)
- Pavel Zhabyeyev
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- MazankowskiAlberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Chandu Sadasivan
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- MazankowskiAlberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Saumya Shah
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- MazankowskiAlberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Faqi Wang
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Gavin Y. Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- MazankowskiAlberta Heart Institute, University of Alberta, Edmonton, AB, Canada
- Correspondence: Gavin Y. Oudit
| |
Collapse
|
41
|
Zhang J, Song Y, Li Y, Lin HB, Fang X. Iron homeostasis in the heart: Molecular mechanisms and pharmacological implications. J Mol Cell Cardiol 2023; 174:15-24. [PMID: 36375319 DOI: 10.1016/j.yjmcc.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022]
Abstract
Iron is necessary for the life of practically all living things, yet it may also harm people toxically. Accordingly, humans and other mammals have evolved an effective and tightly regulatory system to maintain iron homeostasis in healthy tissues, including the heart. Iron deficiency is common in patients with heart failure, and is associated with worse prognosis in this population; while the prevalence of iron overload-related cardiovascular disorders is also increasing. Therefore, enhancing the therapy of patients with cardiovascular disorders requires a thorough understanding of iron homeostasis. Here, we give readers an overview of the fundamental mechanisms governing systemic iron homeostasis as well as the most recent knowledge about the intake, storage, use, and export of iron from the heart. Genetic mouse models used for investigation of iron metabolism in various in vivo scenarios are summarized and highlighted. We also go through different clinical conditions and therapeutic approaches that target cardiac iron dyshomeostasis. Finally, we conclude the review by outlining the present knowledge gaps and important open questions in this field in order to guide future research on cardiac iron metabolism.
Collapse
Affiliation(s)
- Jiawei Zhang
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yijing Song
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - You Li
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Han-Bin Lin
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xuexian Fang
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.
| |
Collapse
|
42
|
Qiu F, Wu L, Yang G, Zhang C, Liu X, Sun X, Chen X, Wang N. The role of iron metabolism in chronic diseases related to obesity. Mol Med 2022; 28:130. [PMID: 36335331 PMCID: PMC9636637 DOI: 10.1186/s10020-022-00558-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/14/2022] [Indexed: 11/08/2022] Open
Abstract
Obesity is one of the major public health problems threatening the world, as well as a potential risk factor for chronic metabolic diseases. There is growing evidence that iron metabolism is altered in obese people, however, the highly refined regulation of iron metabolism in obesity and obesity-related complications is still being investigated. Iron accumulation can affect the body’s sensitivity to insulin, Type 2 diabetes, liver disease and cardiovascular disease. This review summarized the changes and potential mechanisms of iron metabolism in several chronic diseases related to obesity, providing new clues for future research.
Collapse
|
43
|
Darvishi‐Khezri H, Khalilzadeh Arjmandi H, Aliasgharian A, Shaki F, Zahedi M, Kosaryan M, Karami H, Naeimayi Aali R, Salehifar E. Amlodipine: Can act as an antioxidant in patients with transfusion-dependent β-thalassemia? A double-blind, controlled, crossover trial. J Clin Lab Anal 2022; 36:e24752. [PMID: 36357338 PMCID: PMC9756999 DOI: 10.1002/jcla.24752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 10/15/2022] [Accepted: 10/21/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND AND AIM This study aimed to assess the antioxidant effects of amlodipine in transfusion-dependent β-thalassemia (TDT) patients. METHODS This crossover trial consisted of two sequences (AP and PA). In the AP sequence, nine cases received amlodipine 5 mg daily (phase I) and then were switched to placebo (phase II). In PA sequence, 10 patients took the placebo (phase I) and were shifted to amlodipine (phase II). The washout period was 2 weeks. The length of each phase was 6 months. Serum malondialdehyde (MDA, μmol/L), carbonyl (protein CO, μM/L), glutathione (GSH, nM/L), and total antioxidant capacity (TAC, μmol FeSO4/L) were measured in the beginning and at the end of phases I and II. The clinical significance was viewed as a minimum change difference of 5% for each outcome between amlodipine and placebo. RESULTS Seventeen cases completed the study. According to the baseline MDA values, the adjusted Hedges's g for MDA was -0.59, 95% confidence interval [CI] -1.26 to 0.08. After controlling the baseline protein CO values, Hedges's g computed for protein CO was -0.11, 95% CI -0.76 to 0.55. The estimated values of the adjusted Hedges's g for GSH and TAC were also 0.26, 95% CI -0.40 to 0.91, and 0.42, 95% CI -0.24 to 1.09, respectively. The change difference for MDA was 8.3% (protein CO 2.2%, GSH 3.1%, and TAC 12.9%). CONCLUSION Clinically, amlodipine therapy is an efficacious adjuvant treatment with conventional iron chelators for improving the levels of MDA and TAC in patients with TDT.
Collapse
Affiliation(s)
- Hadi Darvishi‐Khezri
- Thalassemia Research Center (TRC), Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran
| | - Hadiseh Khalilzadeh Arjmandi
- Student Research Committee, Pharmaceutical Sciences Research Center, Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran
| | - Aily Aliasgharian
- Thalassemia Research Center (TRC), Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran
| | - Fatemeh Shaki
- Department of Toxicology and Pharmacology, Faculty of PharmacyMazandaran University of Medical SciencesSariIran
| | - Mohammad Zahedi
- Thalassemia Research Center (TRC), Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran,Student Research CommitteeSchool of Allied Medicine, Iran University of Medical SciencesTehranIran
| | - Mehrnoush Kosaryan
- Thalassemia Research Center (TRC), Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran
| | - Hossein Karami
- Thalassemia Research Center (TRC), Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran
| | | | - Ebrahim Salehifar
- Pharmaceutical Sciences Research Center, Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran
| |
Collapse
|
44
|
Rapezzi C, Aimo A, Barison A, Emdin M, Porcari A, Linhart A, Keren A, Merlo M, Sinagra G. Restrictive cardiomyopathy: definition and diagnosis. Eur Heart J 2022; 43:4679-4693. [PMID: 36269634 PMCID: PMC9712030 DOI: 10.1093/eurheartj/ehac543] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/23/2022] [Accepted: 09/16/2022] [Indexed: 01/05/2023] Open
Abstract
Restrictive cardiomyopathy (RCM) is a heterogeneous group of diseases characterized by restrictive left ventricular pathophysiology, i.e. a rapid rise in ventricular pressure with only small increases in filling volume due to increased myocardial stiffness. More precisely, the defining feature of RCM is the coexistence of persistent restrictive pathophysiology, diastolic dysfunction, non-dilated ventricles, and atrial dilatation, regardless of ventricular wall thickness and systolic function. Beyond this shared haemodynamic hallmark, the phenotypic spectrum of RCM is wide. The disorders manifesting as RCM may be classified according to four main disease mechanisms: (i) interstitial fibrosis and intrinsic myocardial dysfunction, (ii) infiltration of extracellular spaces, (iii) accumulation of storage material within cardiomyocytes, or (iv) endomyocardial fibrosis. Many disorders do not show restrictive pathophysiology throughout their natural history, but only at an initial stage (with an evolution towards a hypokinetic and dilated phenotype) or at a terminal stage (often progressing from a hypertrophic phenotype). Furthermore, elements of both hypertrophic and restrictive phenotypes may coexist in some patients, making the classification challenge. Restrictive pathophysiology can be demonstrated by cardiac catheterization or Doppler echocardiography. The specific conditions may usually be diagnosed based on clinical data, 12-lead electrocardiogram, echocardiography, nuclear medicine, or cardiovascular magnetic resonance, but further investigations may be needed, up to endomyocardial biopsy and genetic evaluation. The spectrum of therapies is also wide and heterogeneous, but disease-modifying treatments are available only for cardiac amyloidosis and, partially, for iron overload cardiomyopathy.
Collapse
Affiliation(s)
- Claudio Rapezzi
- Corresponding author. Tel: +39 0532239882, Fax: +39 0532 293031,
| | - Alberto Aimo
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, piazza Martiri della Libertà 33, 56127 Pisa, Italy,Cardiology Division, Fondazione Toscana Gabriele Monasterio, via Moruzzi 1, 56124 Pisa, Italy
| | - Andrea Barison
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, piazza Martiri della Libertà 33, 56127 Pisa, Italy,Cardiology Division, Fondazione Toscana Gabriele Monasterio, via Moruzzi 1, 56124 Pisa, Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, piazza Martiri della Libertà 33, 56127 Pisa, Italy,Cardiology Division, Fondazione Toscana Gabriele Monasterio, via Moruzzi 1, 56124 Pisa, Italy
| | - Aldostefano Porcari
- Center for Diagnosis and Treatment of Cardiomyopathies, Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI), University of Trieste, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart, Via Giacomo Puccini, 50, 34148 Trieste, Italy
| | - Ales Linhart
- General University Hospital and Charles University, Opletalova 38, 110 00 Staré Město, Czech Republic
| | - Andre Keren
- Cardiology Division, Hadassah Hebrew University Hospital, Sderot Churchill 8, Jerusalem, Israel,Heart Failure Center, Clalit Health Services, Bnei Brit St 22, Jerusalem, Israel
| | - Marco Merlo
- Center for Diagnosis and Treatment of Cardiomyopathies, Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI), University of Trieste, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart, Via Giacomo Puccini, 50, 34148 Trieste, Italy
| | - Gianfranco Sinagra
- Center for Diagnosis and Treatment of Cardiomyopathies, Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI), University of Trieste, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart, Via Giacomo Puccini, 50, 34148 Trieste, Italy
| |
Collapse
|
45
|
Gwanyanya A, Mubagwa K. Emerging role of transient receptor potential (TRP) ion channels in cardiac fibroblast pathophysiology. Front Physiol 2022; 13:968393. [PMID: 36277180 PMCID: PMC9583832 DOI: 10.3389/fphys.2022.968393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiac fibroblasts make up a major proportion of non-excitable cells in the heart and contribute to the cardiac structural integrity and maintenance of the extracellular matrix. During myocardial injury, fibroblasts can be activated to trans-differentiate into myofibroblasts, which secrete extracellular matrix components as part of healing, but may also induce cardiac fibrosis and pathological cardiac structural and electrical remodeling. The mechanisms regulating such cellular processes still require clarification, but the identification of transient receptor potential (TRP) channels in cardiac fibroblasts could provide further insights into the fibroblast-related pathophysiology. TRP proteins belong to a diverse superfamily, with subgroups such as the canonical (TRPC), vanilloid (TRPV), melastatin (TRPM), ankyrin (TRPA), polycystin (TRPP), and mucolipin (TRPML). Several TRP proteins form non-selective channels that are permeable to cations like Na+ and Ca2+ and are activated by various chemical and physical stimuli. This review highlights the role of TRP channels in cardiac fibroblasts and the possible underlying signaling mechanisms. Changes in the expression or activity of TRPs such as TRPCs, TRPVs, TRPMs, and TRPA channels modulate cardiac fibroblasts and myofibroblasts, especially under pathological conditions. Such TRPs contribute to cardiac fibroblast proliferation and differentiation as well as to disease conditions such as cardiac fibrosis, atrial fibrillation, and fibroblast metal toxicity. Thus, TRP channels in fibroblasts represent potential drug targets in cardiac disease.
Collapse
Affiliation(s)
- Asfree Gwanyanya
- Department of Human Biology, University of Cape Town, Cape Town, South Africa
- *Correspondence: Asfree Gwanyanya,
| | - Kanigula Mubagwa
- Department of Cardiovascular Sciences, K U Leuven, Leuven, Belgium
- Department of Basic Sciences, Faculty of Medicine, Université Catholique de Bukavu, Bukavu, Democratic Republic of Congo
| |
Collapse
|
46
|
Ferroptosis: The Potential Target in Heart Failure with Preserved Ejection Fraction. Cells 2022; 11:cells11182842. [PMID: 36139417 PMCID: PMC9496758 DOI: 10.3390/cells11182842] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 12/01/2022] Open
Abstract
Ferroptosis is a recently identified cell death characterized by an excessive accumulation of iron-dependent reactive oxygen species (ROS) and lipid peroxides. Intracellular iron overload can not only cause damage to macrophages, endothelial cells, and cardiomyocytes through responses such as lipid peroxidation, oxidative stress, and inflammation, but can also affect cardiomyocyte Ca2+ handling, impair excitation–contraction coupling, and play an important role in the pathological process of heart failure with preserved ejection fraction (HFpEF). However, the mechanisms through which ferroptosis initiates the development and progression of HFpEF have not been established. This review explains the possible correlations between HFpEF and ferroptosis and provides a reliable theoretical basis for future studies on its mechanism.
Collapse
|
47
|
Clinical and Molecular Aspects of Iron Metabolism in Failing Myocytes. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081203. [PMID: 36013382 PMCID: PMC9409945 DOI: 10.3390/life12081203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022]
Abstract
Heart failure (HF) is a common disease that causes significant limitations on the organism's capacity and, in extreme cases, leads to death. Clinically, iron deficiency (ID) plays an essential role in heart failure by deteriorating the patient's condition and is a prognostic marker indicating poor clinical outcomes. Therefore, in HF patients, supplementation of iron is recommended. However, iron treatment may cause adverse effects by increasing iron-related apoptosis and the production of oxygen radicals, which may cause additional heart damage. Furthermore, many knowledge gaps exist regarding the complex interplay between iron deficiency and heart failure. Here, we describe the current, comprehensive knowledge about the role of the proteins involved in iron metabolism. We will focus on the molecular and clinical aspects of iron deficiency in HF. We believe that summarizing the new advances in the translational and clinical research regarding iron deficiency in heart failure should broaden clinicians' awareness of this comorbidity.
Collapse
|
48
|
Li D, Pi W, Sun Z, Liu X, Jiang J. Ferroptosis and its role in cardiomyopathy. Biomed Pharmacother 2022; 153:113279. [PMID: 35738177 DOI: 10.1016/j.biopha.2022.113279] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 12/09/2022] Open
Abstract
Heart disease is the leading cause of death worldwide. Cardiomyopathy is a disease characterized by the heart muscle damage, resulting heart in a structurally and functionally change, as well as heart failure and sudden cardiac death. The key pathogenic factor of cardiomyopathy is the loss of cardiomyocytes, but the related molecular mechanisms remain unclear. Ferroptosis is a newly discovered regulated form of cell death, characterized by iron accumulation and lipid peroxidation during cell death. Recent studies have shown that ferroptosis plays an important regulatory roles in the occurrence and development of many heart diseases such as myocardial ischemia/reperfusion injury, cardiomyopathy and heart failure. However, the systemic association of ferroptosis and cardiomyopathy remains largely unknown and needs to be elucidated. In this review, we provide an overview of the molecular mechanisms of ferroptosis and its role in individual cardiomyopathies, highlight that targeting ferroptosis maybe a potential therapeutic strategy for cardiomyopathy therapy in the future.
Collapse
Affiliation(s)
- Danlei Li
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Zhenzhu Sun
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Xiaoman Liu
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China.
| |
Collapse
|
49
|
Zhang H, Jamieson KL, Grenier J, Nikhanj A, Tang Z, Wang F, Wang S, Seidman JG, Seidman CE, Thompson R, Seubert JM, Oudit GY. Myocardial Iron Deficiency and Mitochondrial Dysfunction in Advanced Heart Failure in Humans. J Am Heart Assoc 2022; 11:e022853. [PMID: 35656974 PMCID: PMC9238720 DOI: 10.1161/jaha.121.022853] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Myocardial iron deficiency (MID) in heart failure (HF) remains largely unexplored. We aim to establish defining criterion for MID, evaluate its pathophysiological role, and evaluate the applicability of monitoring it non‐invasively in human explanted hearts. Methods and Results Biventricular tissue iron levels were measured in both failing (n=138) and non‐failing control (NFC, n=46) explanted human hearts. Clinical phenotyping was complemented with comprehensive assessment of myocardial remodeling and mitochondrial functional profiles, including metabolic and oxidative stress. Myocardial iron status was further investigated by cardiac magnetic resonance imaging. Myocardial iron content in the left ventricle was lower in HF versus NFC (121.4 [88.1–150.3] versus 137.4 [109.2–165.9] μg/g dry weight), which was absent in the right ventricle. With a priori cutoff of 86.1 μg/g d.w. in left ventricle, we identified 23% of HF patients with MID (HF‐MID) associated with higher NYHA class and worsened left ventricle function. Respiratory chain and Krebs cycle enzymatic activities were suppressed and strongly correlated with depleted iron stores in HF‐MID hearts. Defenses against oxidative stress were severely impaired in association with worsened adverse remodeling in iron‐deficient hearts. Mechanistically, iron uptake pathways were impeded in HF‐MID including decreased translocation to the sarcolemma, while transmembrane fraction of ferroportin positively correlated with MID. Cardiac magnetic resonance with T2* effectively captured myocardial iron levels in failing hearts. Conclusions MID is highly prevalent in advanced human HF and exacerbates pathological remodeling in HF driven primarily by dysfunctional mitochondria and increased oxidative stress in the left ventricle. Cardiac magnetic resonance demonstrates clinical potential to non‐invasively monitor MID.
Collapse
Affiliation(s)
- Hao Zhang
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - K Lockhart Jamieson
- Department of Pharmacology Faculty of Medicine and DentistryEdmonton Alberta Canada
| | - Justin Grenier
- Mazankowski Alberta Heart Institute Edmonton Alberta Canada.,Department of Biomedical Engineering Faculty of Medicine and DentistryEdmonton Alberta Canada
| | - Anish Nikhanj
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Zeyu Tang
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Faqi Wang
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Shaohua Wang
- Mazankowski Alberta Heart Institute Edmonton Alberta Canada.,Division of Cardiac Surgery Department of Surgery Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | | | - Christine E Seidman
- Department of Genetics Harvard Medical School Boston MA.,Cardiovascular Division Brigham and Women's Hospital Boston MA
| | - Richard Thompson
- Mazankowski Alberta Heart Institute Edmonton Alberta Canada.,Department of Biomedical Engineering Faculty of Medicine and DentistryEdmonton Alberta Canada
| | - John M Seubert
- Mazankowski Alberta Heart Institute Edmonton Alberta Canada.,Department of Pharmacology Faculty of Medicine and DentistryEdmonton Alberta Canada
| | - Gavin Y Oudit
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| |
Collapse
|
50
|
Berezovsky B, Frýdlová J, Gurieva I, Rogalsky DW, Vokurka M, Krijt J. Heart Ferroportin Protein Content Is Regulated by Heart Iron Concentration and Systemic Hepcidin Expression. Int J Mol Sci 2022; 23:ijms23115899. [PMID: 35682577 PMCID: PMC9180074 DOI: 10.3390/ijms23115899] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 02/06/2023] Open
Abstract
The purpose of the study was to investigate the expression of ferroportin protein following treatments that affect systemic hepcidin. Administration of erythropoietin to C57BL/6J mice decreased systemic hepcidin expression; it also increased heart ferroportin protein content, determined by immunoblot in the membrane fraction, to approximately 200% of control values. This increase in heart ferroportin protein is very probably caused by a decrease in systemic hepcidin expression, in accordance with the classical regulation of ferroportin by hepcidin. However, the control of heart ferroportin protein by systemic hepcidin could apparently be overridden by changes in heart non-heme iron content since injection of ferric carboxymaltose to mice at 300 mg Fe/kg resulted in an increase in liver hepcidin expression, heart non-heme iron content, and also a threefold increase in heart ferroportin protein content. In a separate experiment, feeding an iron-deficient diet to young Wistar rats dramatically decreased liver hepcidin expression, while heart non-heme iron content and heart ferroportin protein content decreased to 50% of controls. It is, therefore, suggested that heart ferroportin protein is regulated primarily by the iron regulatory protein/iron-responsive element system and that the regulation of heart ferroportin by the hepcidin-ferroportin axis plays a secondary role.
Collapse
Affiliation(s)
- Betty Berezovsky
- Institute of Pathophysiology, First Faculty of Medicine, Charles University, 128 53 Prague, Czech Republic; (B.B.); (J.F.); (I.G.); (M.V.)
| | - Jana Frýdlová
- Institute of Pathophysiology, First Faculty of Medicine, Charles University, 128 53 Prague, Czech Republic; (B.B.); (J.F.); (I.G.); (M.V.)
| | - Iuliia Gurieva
- Institute of Pathophysiology, First Faculty of Medicine, Charles University, 128 53 Prague, Czech Republic; (B.B.); (J.F.); (I.G.); (M.V.)
| | | | - Martin Vokurka
- Institute of Pathophysiology, First Faculty of Medicine, Charles University, 128 53 Prague, Czech Republic; (B.B.); (J.F.); (I.G.); (M.V.)
| | - Jan Krijt
- Institute of Pathophysiology, First Faculty of Medicine, Charles University, 128 53 Prague, Czech Republic; (B.B.); (J.F.); (I.G.); (M.V.)
- Correspondence:
| |
Collapse
|