101
|
Tian Y, Li M, Cheng R, Chen X, Xu Z, Yuan J, Diao Z, Hao L. Human adipose mesenchymal stem cell-derived exosomes alleviate fibrosis by restraining ferroptosis in keloids. Front Pharmacol 2024; 15:1431846. [PMID: 39221144 PMCID: PMC11361945 DOI: 10.3389/fphar.2024.1431846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
Background Keloid is a fibroproliferative disease with unsatisfactory therapeutic effects and a high recurrence rate. exosomes produced by adipose-derived mesenchymal stem cells (ADSC-Exos) have attracted significant interest due to their ability to treat fibrosis. However, the molecular mechanisms of ADSC-Exos in keloids remain inconclusive. Objective Our study revealed the relationship between ferroptosis and fibrosis in keloids. Subsequently, this study aimed to explore further the anti-fibrotic effect of ADSC-Exos on keloids through ferroptosis and the potential underlying mechanisms. Methods To investigate the impact of ferroptosis on keloid fibrosis, Erastin and ferrostatin-1 (fer-1) were utilized to treat keloid fibroblast. Keloid keloids treated with Erastin and fer-1 were cocultured with ADSC-Exos to validate the impact of ferroptosis on the effect of ADSC-Exos on keloid anti-ferrotic protein, peroxidase 4 (GPX4) and anti-fibrotic effects in vivo and in vitro by Western blot, as well as variations in iron metabolite expression, malondialdehyde (MDA), liposomal peroxidation (LPO) and glutathione (GSH) were analyzed. The effect of solute carrier family 7-member 11 (SLC7A11) silencing on ADSC-Exo-treated keloid fibroblast was investigated. Results Iron metabolite dysregulation was validated in keloids. Fibrosis progression is enhanced by Erastin-induced ferroptosis. The anti-fibrotic effects of ADSC-Exos and fer-1 are related to their ability to prevent iron metabolism. ADSC-Exos effectively suppressed keloid fibrosis progression and increased GSH and GPX4 gene expression. Additionally, the use of Erastin limits the effect of ADSC-Exos in keloids. Furthermore, the effect of ADSC-Exos on keloids was associated with SLC7A11-GPX4 signaling pathway. Conclusion We demonstrated a new potential mechanism by which anti-ferroptosis inhibits the progression of keloid fibrosis and identified an ADSC-Exo-based keloid therapeutic strategy. Resisting the occurrence of ferroptosis and the existence of the SLC7A11-GPX4 signaling pathway might serve as a target for ADSC-Exos.
Collapse
|
102
|
Chen Y, Cui Y, Li M, Xia M, Xiang Q, Mao Y, Li H, Chen J, Zeng W, Zheng X, Peng J, Dai X, Tang Z. A novel mechanism of ferroptosis inhibition-enhanced atherosclerotic plaque stability: YAP1 suppresses vascular smooth muscle cell ferroptosis through GLS1. FASEB J 2024; 38:e23850. [PMID: 39091212 DOI: 10.1096/fj.202401251r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/05/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Abstract
Atherosclerosis is a leading cause of cardiovascular diseases (CVDs), often resulting in major adverse cardiovascular events (MACEs), such as myocardial infarction and stroke due to the rupture or erosion of vulnerable plaques. Ferroptosis, an iron-dependent form of cell death, has been implicated in the development of atherosclerosis. Despite its involvement in CVDs, the specific role of ferroptosis in atherosclerotic plaque stability remains unclear. In this study, we confirmed the presence of ferroptosis in unstable atherosclerotic plaques and demonstrated that the ferroptosis inhibitor ferrostatin-1 (Fer-1) stabilizes atherosclerotic plaques in apolipoprotein E knockout (Apoe-/-) mice. Using bioinformatic analysis combining RNA sequencing (RNA-seq) with single-cell RNA sequencing (scRNA-seq), we identified Yes-associated protein 1 (YAP1) as a potential key regulator of ferroptosis in vascular smooth muscle cells (VSMCs) of unstable plaques. In vitro, we found that YAP1 protects against oxidized low-density lipoprotein (oxLDL)-induced ferroptosis in VSMCs. Mechanistically, YAP1 exerts its anti-ferroptosis effects by regulating the expression of glutaminase 1 (GLS1) to promote the synthesis of glutamate (Glu) and glutathione (GSH). These findings establish a novel mechanism where the inhibition of ferroptosis promotes the stabilization of atherosclerotic plaques through the YAP1/GLS1 axis, attenuating VSMC ferroptosis. Thus, targeting the YAP1/GLS1 axis to suppress VSMC ferroptosis may represent a novel strategy for preventing and treating unstable atherosclerotic plaques.
Collapse
MESH Headings
- Ferroptosis
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- YAP-Signaling Proteins/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Humans
- Male
- Mice, Inbred C57BL
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Mice, Knockout
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Phenylenediamines/pharmacology
- Cyclohexylamines/pharmacology
- Apolipoproteins E/metabolism
- Apolipoproteins E/genetics
Collapse
Affiliation(s)
- Yanyu Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Yuting Cui
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Man Li
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Mengdie Xia
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Qiong Xiang
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Yu Mao
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Hengjuan Li
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Jialin Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Wen Zeng
- Shaoyang Branch of Key Laboratory for Arteriosclerology of Hunan Province, The Central Hospital of Shaoyang, Shaoyang, China
| | - Xilong Zheng
- Department of Biochemistry & Molecular Biology and Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Juan Peng
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
- Shaoyang Branch of Key Laboratory for Arteriosclerology of Hunan Province, The Central Hospital of Shaoyang, Shaoyang, China
| | - Xiaoyan Dai
- Clinical Research Institute, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhihan Tang
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
- Shaoyang Branch of Key Laboratory for Arteriosclerology of Hunan Province, The Central Hospital of Shaoyang, Shaoyang, China
| |
Collapse
|
103
|
Chen X, Zhang X, Wu Y, Wang Z, Yu T, Chen P, Tong P, Gao J, Chen H. The Iron Binding Ability Maps the Fate of Food-Derived Transferrins: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:17771-17781. [PMID: 39087686 DOI: 10.1021/acs.jafc.4c04827] [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: 08/02/2024]
Abstract
As the demand for lactoferrin increases, the search for cost-effective alternative proteins becomes increasingly important. Attention naturally turns to other members of the transferrin family such as ovotransferrin. The iron-binding abilities of these proteins influence their characteristics, although the underlying mechanisms remain unclear. This overview systematically summarizes the effects of the iron-binding ability on the fate of food-derived transferrins (lactoferrin and ovotransferrin) and their potential applications. The findings indicate that iron-binding ability significantly influences the structure of food-derived transferrins, particularly their tertiary structure. Changes in structure influence their physicochemical properties, which, in turn, lead to different behaviors in response to environmental variations. Thus, these proteins exhibit distinct digestive characteristics by the time they reach the small intestine, ultimately performing varied physiological functions in vivo. Consequently, food-derived transferrins with different iron-binding states may find diverse applications. Understanding this capability is essential for developing food-derived transferrins and driving innovation in lactoferrin-related industries.
Collapse
Affiliation(s)
- Xiao Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- College of Food Science and Technology, Nanchang University, Nanchang 330031, China
- Sino German Joint Research Institute, Nanchang University, Nanchang 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Xing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- College of Food Science and Technology, Nanchang University, Nanchang 330031, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Yong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- Sino German Joint Research Institute, Nanchang University, Nanchang 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Zhongliang Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- College of Food Science and Technology, Nanchang University, Nanchang 330031, China
- Sino German Joint Research Institute, Nanchang University, Nanchang 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Tian Yu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- College of Food Science and Technology, Nanchang University, Nanchang 330031, China
- Sino German Joint Research Institute, Nanchang University, Nanchang 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Pingduo Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- College of Food Science and Technology, Nanchang University, Nanchang 330031, China
- Sino German Joint Research Institute, Nanchang University, Nanchang 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Ping Tong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- Sino German Joint Research Institute, Nanchang University, Nanchang 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- College of Food Science and Technology, Nanchang University, Nanchang 330031, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- Sino German Joint Research Institute, Nanchang University, Nanchang 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| |
Collapse
|
104
|
Wu L, Wang LT, Du YX, Zhang YM, Ren J. Asiatic acid ameliorates doxorubicin-induced cardiotoxicity by promoting FPN-mediated iron export and inhibiting ferroptosis. Acta Pharmacol Sin 2024:10.1038/s41401-024-01367-9. [PMID: 39143234 DOI: 10.1038/s41401-024-01367-9] [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: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 08/16/2024] Open
Abstract
Doxorubicin (DOX), a common chemotherapeutic agent in cancer therapy, is accompanied by pronounced cardiotoxicity. Ferroptosis has been implicated in the pathogenesis and therapeutics of DOX-induced cardiotoxicity (DIC). Asiatic acid (AA), a pentacyclic triterpene from the Chinese medicinal herb Centella asiatica, displays antioxidant, anti-inflammatory, and antiapoptotic activities. In this study, we investigated the beneficial effects of AA against DOX-induced ferroptosis and cardiotoxicity and the underlying mechanisms. A chronic DIC model was established by challenging mice with DOX (5 mg/kg, i.p.) once per week for 4 weeks. Concurrent with DOX insult, the mice were administered AA (25 mg·kg-1·d-1, i.g.). Cardiac function and mechanical properties of isolated cardiomyocytes were evaluated at the end of treatment. We showed that AA administration preserved cardiac function, significantly reduced cardiac injury, and improved cardiomyocyte contractile function in DIC mice. The beneficial effects of AA were causally linked to the inhibition of DOX-induced ferroptosis both in vivo and in vitro. We revealed that AA attenuated DOX-induced iron accumulation in HL-1 cells by increasing FPN-mediated iron export, in a Nrf2-dependent manner. AA upregulated Nrf2 expression and promoted Nrf2 nuclear translocation in DOX-treated HL-1 cells. Moreover, AA-offered benefits against DOX-induced cardiac dysfunction and ferroptosis were abolished by Nrf2 inhibitor ML385 (30 mg·kg-1·d-1, i.p.) administrated 30 min before AA in DIC mice. Our data favor that AA promotes FPN-mediated iron export to inhibit iron overload and ferroptosis in DIC, suggesting its therapeutic potential in the treatment of DIC.
Collapse
Affiliation(s)
- 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
| | - Li-Tao Wang
- 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
| | - Yu-Xin Du
- 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
| | - Ying-Mei Zhang
- 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.
| | - Jun Ren
- 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.
| |
Collapse
|
105
|
Li Y, Wu W, Song Y, Zhang J, Han D, Shu C, Lian F, Fang X. β-Caryophyllene Confers Cardioprotection by Scavenging Radicals and Blocking Ferroptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18003-18012. [PMID: 39088660 DOI: 10.1021/acs.jafc.4c03239] [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: 08/03/2024]
Abstract
Ferroptosis is a form of regulated cell death triggered by iron-dependent lipid peroxidation and has been associated with heart diseases. However, there are currently no approved drugs that specifically inhibit ferroptosis in clinical practice, which largely limits the translational potential of this novel target. Here, we demonstrated that β-caryophyllene (BCP; 150 μM), a natural dietary cannabinoid, protects cardiomyocytes against ferroptotic cell death induced by cysteine deprivation or glutathione peroxidase 4 (GPX4) inactivation. Moreover, BCP preserved the mitochondrial morphology and function during ferroptosis induction. Unexpectedly, BCP supported ferroptosis resistance independent of canonical antiferroptotic pathways. Our results further suggested that BCP may terminate radical chain reactions through interactions with molecular oxygen, which also explains why its oxidation derivative failed to suppress ferroptosis. Finally, oral BCP administration (50 mg/kg, daily) significantly alleviated doxorubicin (15 mg/kg, single i.p. injection)-induced cardiac ferroptosis and cardiomyopathy in mice. In conclusion, our data revealed the role of BCP as a natural antiferroptotic compound and suggest pharmacological modification based on BCP as a promising therapeutic strategy for treating ferroptosis-associated heart disorders.
Collapse
Affiliation(s)
- You Li
- School of Public Health, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Wei Wu
- School of Public Health, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yijing Song
- School of Public Health, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jiawei Zhang
- School of Public Health, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Dan Han
- Department of Nutrition and Food Safety, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang 310051, China
| | - Chi Shu
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Fuzhi Lian
- School of Public Health, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xuexian Fang
- School of Public Health, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
106
|
Zhao Y, Fei Y, Zhao Y, Li M, Hu Y, Cai K, Yu SH, Luo Z. Biomineralization-Tuned Nanounits Reprogram the Signal Transducer and Activator of Transcription 3 Signaling for Ferroptosis-Immunotherapy in Cancer Stem Cells. ACS NANO 2024; 18:21268-21287. [PMID: 39083438 DOI: 10.1021/acsnano.4c05084] [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: 08/02/2024]
Abstract
Cancer stem cells (CSCs) are promising targets for improving anticancer treatment outcomes while eliminating recurrence, but their treatment remains a major challenge. Here, we report a nanointegrative strategy to realize CSC-targeted ferroptosis-immunotherapy through spatiotemporally controlled reprogramming of STAT3-regulated signaling circuits. Specifically, STAT3 inhibitor niclosamide (Ni) and an experimental ferroptosis drug (1S, 3R)-RSL3 (RSL3) are integrated into hyaluronic acid-modified amorphous calcium phosphate (ACP) nanounits through biomineralization (CaP-PEG-HA@Ni/RSL3), which could be recognized by CD44-overexpressing CSCs and released in a synchronized manner. Ni inhibits the CSC-intrinsic STAT3-PD-L1 axis to stimulate adaptive immunity and enhance interferon gamma (IFNγ) secretion by CD8+ T cells to downregulate SLC7A11 and SLC3A2 for blocking glutathione biosynthesis. Meanwhile, Ni-dependent STAT3 inhibition also upregulates ACSL4 through downstream signaling and IFNγ feedback. These effects cooperate with RSL3-mediated GPX4 deactivation to induce pronounced ferroptosis. Furthermore, CaP-PEG-HA@Ni/RSL3 also impairs the immunosuppressive M2-like tumor-associated macrophages, while Ca2+ ions released from degraded ACP could chelate with lipid peroxides in ferroptotic CSCs to avoid CD8+ T-cell inhibition, thus boosting the effector function of activated CD8+ T cells. This study offers a cooperative ferroptosis-immunotherapeutic approach for the treatment of refractory cancer.
Collapse
Affiliation(s)
- Youbo Zhao
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory for Autoimmune Disease Research, Department of Hepatic-Biliary-Pancreatic Surgery Affiliate Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550025, P. R. China
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yang Fei
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yang Zhao
- Division of Nanomaterials and Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Collaborative Innovation Center of Suzhou Nano Science and Technology, Department of Chemistry, Hefei Science Center of CAS, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Shu-Hong Yu
- Division of Nanomaterials and Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Collaborative Innovation Center of Suzhou Nano Science and Technology, Department of Chemistry, Hefei Science Center of CAS, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
107
|
Letafati A, Taghiabadi Z, Ardekani OS, Abbasi S, Najafabadi AQ, Jazi NN, Soheili R, Rodrigo R, Yavarian J, Saso L. Unveiling the intersection: ferroptosis in influenza virus infection. Virol J 2024; 21:185. [PMID: 39135112 PMCID: PMC11321227 DOI: 10.1186/s12985-024-02462-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
The influenza virus (IFV) imposes a considerable health and economic burden globally, requiring a comprehensive understanding of its pathogenic mechanisms. Ferroptosis, an iron-dependent lipid peroxidation cell death pathway, holds unique implications for the antioxidant defense system, with possible contributions to inflammation. This exploration focuses on the dynamic interplay between ferroptosis and the host defense against viruses, emphasizing the influence of IFV infections on the activation of the ferroptosis pathway. IFV causes different types of cell death, including apoptosis, necrosis, and ferroptosis. IFV-induced ferroptotic cell death is mediated by alterations in iron homeostasis, intensifying the accumulation of reactive oxygen species and promoting lipid peroxidation. A comprehensive investigation into the mechanism of ferroptosis in viral infections, specifically IFV, has great potential to identify therapeutic strategies. This understanding may pave the way for the development of drugs using ferroptosis inhibitors, presenting an effective approach to suppress viral infections.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Taghiabadi
- Department of Microbiology and Virology of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Omid Salahi Ardekani
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Simin Abbasi
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Qaraee Najafabadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Negar Nayerain Jazi
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roben Soheili
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jila Yavarian
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University, Rome, Italy.
| |
Collapse
|
108
|
Luo M, Luan X, Yang C, Chen X, Yuan S, Cao Y, Zhang J, Xie J, Luo Q, Chen L, Li S, Xiang W, Zhou J. Revisiting the potential of regulated cell death in glioma treatment: a focus on autophagy-dependent cell death, anoikis, ferroptosis, cuproptosis, pyroptosis, immunogenic cell death, and the crosstalk between them. Front Oncol 2024; 14:1397863. [PMID: 39184045 PMCID: PMC11341384 DOI: 10.3389/fonc.2024.1397863] [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: 03/08/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Gliomas are primary tumors that originate in the central nervous system. The conventional treatment options for gliomas typically encompass surgical resection and temozolomide (TMZ) chemotherapy. However, despite aggressive interventions, the median survival for glioma patients is merely about 14.6 months. Consequently, there is an urgent necessity to explore innovative therapeutic strategies for treating glioma. The foundational study of regulated cell death (RCD) can be traced back to Karl Vogt's seminal observations of cellular demise in toads, which were documented in 1842. In the past decade, the Nomenclature Committee on Cell Death (NCCD) has systematically classified and delineated various forms and mechanisms of cell death, synthesizing morphological, biochemical, and functional characteristics. Cell death primarily manifests in two forms: accidental cell death (ACD), which is caused by external factors such as physical, chemical, or mechanical disruptions; and RCD, a gene-directed intrinsic process that coordinates an orderly cellular demise in response to both physiological and pathological cues. Advancements in our understanding of RCD have shed light on the manipulation of cell death modulation - either through induction or suppression - as a potentially groundbreaking approach in oncology, holding significant promise. However, obstacles persist at the interface of research and clinical application, with significant impediments encountered in translating to therapeutic modalities. It is increasingly apparent that an integrative examination of the molecular underpinnings of cell death is imperative for advancing the field, particularly within the framework of inter-pathway functional synergy. In this review, we provide an overview of various forms of RCD, including autophagy-dependent cell death, anoikis, ferroptosis, cuproptosis, pyroptosis and immunogenic cell death. We summarize the latest advancements in understanding the molecular mechanisms that regulate RCD in glioma and explore the interconnections between different cell death processes. By comprehending these connections and developing targeted strategies, we have the potential to enhance glioma therapy through manipulation of RCD.
Collapse
Affiliation(s)
- Maowen Luo
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xingzhao Luan
- Department of Neurosurgery, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Chaoge Yang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Xiaofan Chen
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Suxin Yuan
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Youlin Cao
- Department of Neurosurgery, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Jing Zhang
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Jiaying Xie
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Qinglian Luo
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Shenjie Li
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Wei Xiang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Jie Zhou
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| |
Collapse
|
109
|
Xu L, Li W, Chen Y, Liu S, Liu G, Luo W, Cao G, Wang S. Metformin Regulates Cardiac Ferroptosis to Reduce Metabolic Syndrome-Induced Cardiac Dysfunction. Appl Biochem Biotechnol 2024:10.1007/s12010-024-05038-7. [PMID: 39106027 DOI: 10.1007/s12010-024-05038-7] [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] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
High-fat diet-induced metabolic syndrome (MetS) is closely associated with cardiac dysfunction. Recent research studies have indicated a potential association between MetS and ferroptosis. Furthermore, metformin can alleviate MetS-induced cardiac ferroptosis. Metformin is a classic biguanide anti-diabetic drug that has protective effects on cardiovascular diseases, which extend beyond its indirect glycemic control. This study aimed to assess whether MetS mediates cardiac ferroptosis, thereby causing oxidative stress and mitochondrial dysfunction. The results revealed that metformin can mitigate cardiac reactive oxygen species and mitochondrial damage, thereby preserving cardiac function. Mechanistic analysis revealed that metformin upregulates the expression of cardiac Nrf2. Moreover, Nrf2 downregulation compromises the cardio-protective effects of metformin. In summary, this study indicated that MetS promotes cardiac ferroptosis, and metformin plays a preventive and therapeutic role, partially through modulation of Nrf2 expression.
Collapse
Affiliation(s)
- Liancheng Xu
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
- Fujian Medical University, Fuzhou, 350108, China
| | - Wenwen Li
- Department of Nephrology, Suqian First Hospital, Suqian, 223800, China
| | - Yu Chen
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Shan Liu
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Guodong Liu
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Weihuan Luo
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Guanyi Cao
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Shiping Wang
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China.
| |
Collapse
|
110
|
Xie Q, Wang J, Li R, Liu H, Zhong Y, Xu Q, Ge Y, Li C, Sun L, Zhu J. IL-6 signaling accelerates iron overload by upregulating DMT1 in endothelial cells to promote aortic dissection. Int J Biol Sci 2024; 20:4222-4237. [PMID: 39247821 PMCID: PMC11379073 DOI: 10.7150/ijbs.99511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/22/2024] [Indexed: 09/10/2024] Open
Abstract
Aortic dissection (AD), caused by tearing of the intima and avulsion of the aortic media, is a severe threat to patient life and organ function. Iron is closely related to dissection formation and organ injury, but the mechanism of iron ion transport disorder in endothelial cells (ECs) remains unclear. We identified the characteristic EC of dissection with iron overload by single-cell RNA sequencing data. After intersecting iron homeostasis and differentially expressed genes, it was found that hypoxia-inducible factor-1α (HIF-1α) and divalent metal transporter 1 (DMT1) are key genes for iron ion disorder. Subsequently, IL-6R was identified as an essential reason for the JAK-STAT activation, a classical iron regulation pathway, through further intersection and validation. In in vivo and in vitro, both high IL-6 receptor expression and elevated IL-6 levels promote JAK1-STAT3 phosphorylation, leading to increased HIF-1α protein levels. Elevated HIF-1α binds explicitly to the 5'-UTR sequence of the DMT1 gene and transcriptionally promotes DMT1 expression, thereby increasing Fe2+ accumulation and endoplasmic reticulum stress (ERS). Blocking IL-6R and free iron with deferoxamine and tocilizumab significantly prolonged survival and reduced aortic and organ damage in dissection mice. A comparison of perioperative data between AD patients and others revealed that high free iron, IL-6, and ERS levels are characteristics of AD patients and are correlated with prognosis. In conclusion, activated IL-6/JAK1/STAT3 signaling axis up-regulates DMT1 expression by increasing HIF-1α, thereby increasing intracellular Fe2+ accumulation and tissue injury, which suggests a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Qiang Xie
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianji Wang
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Runqiao Li
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Hao Liu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yongliang Zhong
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qinfeng Xu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yipeng Ge
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Chengnan Li
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Lizhong Sun
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Junming Zhu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
111
|
Le J, Meng Y, Wang Y, Li D, Zeng F, Xiong Y, Chen X, Deng G. Molecular and therapeutic landscape of ferroptosis in skin diseases. Chin Med J (Engl) 2024; 137:1777-1789. [PMID: 38973265 DOI: 10.1097/cm9.0000000000003164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Indexed: 07/09/2024] Open
Abstract
ABSTRACT Regulated cell death (RCD) is a critical physiological process essential in maintaining skin homeostasis. Among the various forms of RCD, ferroptosis stands out due to its distinct features of iron accumulation, lipid peroxidation, and involvement of various inhibitory antioxidant systems. In recent years, an expanding body of research has solidly linked ferroptosis to the emergence of skin disorders. Therefore, understanding the mechanisms underlying ferroptosis in skin diseases is crucial for advancing therapy and prevention strategies. This review commences with a succinct elucidation of the mechanisms that underpin ferroptosis, embarks on a thorough exploration of ferroptosis's role across a spectrum of skin conditions, encompassing melanoma, psoriasis, systemic lupus erythematosus (SLE), vitiligo, and dermatological ailments precipitated by ultraviolet (UV) exposure, and scrutinizes the potential therapeutic benefits of pharmacological interventions aimed at modulating ferroptosis for the amelioration of skin diseases.
Collapse
Affiliation(s)
- Jiayuan Le
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China
- Furong Laboratory, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China
- Furong Laboratory, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Ying Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China
- Furong Laboratory, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China
- Furong Laboratory, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yixiao Xiong
- Department of Dermatology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China
- Furong Laboratory, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China
- Furong Laboratory, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| |
Collapse
|
112
|
Wang P, Guo X, Wang H, Wang L, Ma M, Guo B. Neuregulin-4 protects cardiomyocytes against high-glucose-induced ferroptosis via the AMPK/NRF2 signalling pathway. Biol Direct 2024; 19:62. [PMID: 39095871 PMCID: PMC11295585 DOI: 10.1186/s13062-024-00505-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND High glucose levels are key factors and key contributors to several cardiovascular diseases associated with cardiomyocyte injury. Ferroptosis, which was identified in recent years, is a mode of cell death caused by the iron-mediated accumulation of lipid peroxides. Neuregulin-4 (Nrg4) is an adipokine that has protective effects against metabolic disorders and insulin resistance. Our previous study revealed that Nrg4 has a protective effect against diabetic myocardial injury, and the aim of this study was to investigate whether Nrg4 could attenuate the occurrence of high glucose-induced ferroptosis in cardiomyocytes. METHODS We constructed an in vivo diabetic myocardial injury model in which primary cardiomyocytes were cultured in vitro and treated with Nrg4. Changes in ferroptosis-related protein levels and ferroptosis-related indices in cardiomyocytes were observed. In addition, we performed back-validation and explored signalling pathways that regulate ferroptosis in primary cardiomyocytes. RESULTS Nrg4 attenuated cardiomyocyte ferroptosis both in vivo and in vitro. Additionally, the AMPK/NRF2 signalling pathway was activated during this process, and when the AMPK/NRF2 pathway was inhibited, the beneficial effects of Nrg4 were attenuated. CONCLUSION Nrg4 antagonizes high glucose-induced ferroptosis in cardiomyocytes via the AMPK/NRF2 signalling pathway.
Collapse
Affiliation(s)
- Pengfei Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
| | - Xiaohua Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
| | - Hongchao Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
| | - Lijie Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
| | - Meifang Ma
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
- Handan Central Hospital, Handan, 056000, China
| | - Bingyan Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China.
- Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
113
|
Cai K, Jiang H, Zou Y, Song C, Cao K, Chen S, Wu Y, Zhang Z, Geng D, Zhang N, Liu B, Sun G, Tang M, Li Z, Zhang Y, Sun Y, Zhang Y. Programmed death of cardiomyocytes in cardiovascular disease and new therapeutic approaches. Pharmacol Res 2024; 206:107281. [PMID: 38942341 DOI: 10.1016/j.phrs.2024.107281] [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: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Cardiovascular diseases (CVDs) have a complex pathogenesis and pose a major threat to human health. Cardiomyocytes have a low regenerative capacity, and their death is a key factor in the morbidity and mortality of many CVDs. Cardiomyocyte death can be regulated by specific signaling pathways known as programmed cell death (PCD), including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, etc. Abnormalities in PCD can lead to the development of a variety of cardiovascular diseases, and there are also molecular-level interconnections between different PCD pathways under the same cardiovascular disease model. Currently, the link between programmed cell death in cardiomyocytes and cardiovascular disease is not fully understood. This review describes the molecular mechanisms of programmed death and the impact of cardiomyocyte death on cardiovascular disease development. Emphasis is placed on a summary of drugs and potential therapeutic approaches that can be used to treat cardiovascular disease by targeting and blocking programmed cell death in cardiomyocytes.
Collapse
Affiliation(s)
- Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Haoyue Jiang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China
| | - Bo Liu
- The first hospital of China Medical University, Department of cardiac surgery, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Man Tang
- Department of clinical pharmacology, College of Pharmacy, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| |
Collapse
|
114
|
Hu K, Jiang P, Hu J, Song B, Hou Y, Zhao J, Chen H, Xie J. Dapagliflozin attenuates LPS-induced myocardial injury by reducing ferroptosis. J Bioenerg Biomembr 2024; 56:361-371. [PMID: 38743190 DOI: 10.1007/s10863-024-10020-3] [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: 03/08/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024]
Abstract
Septic cardiomyopathy is a severe cardiovascular disease with a poor prognosis. Previous studies have reported the involvement of ferroptosis in the pathogenesis of septic cardiomyopathy. SGLT2 inhibitors such as dapagliflozin have been demonstrated to improve ischemia-reperfusion injury by alleviating ferroptosis in cardiomyocyte. However, the role of dapagliflozin in sepsis remains unclear. Therefore, our study aims to investigate the therapeutic effects of dapagliflozin on LPS-induced septic cardiomyopathy. Our results indicate that dapagliflozin improved cardiac function in septic cardiomyopathy experimental mice. Mechanistically, dapagliflozin works by inhibiting the translation of key proteins involved in ferroptosis, such as GPX4, FTH1, and SLC7A11. It also reduces the transcription of lipid peroxidation-related mRNAs, including PTGS2 and ACSL4, as well as iron metabolism genes TFRC and HMOX1.
Collapse
Affiliation(s)
- Ke Hu
- The Affiliated Drum Tower Hospital of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
| | - Pin Jiang
- Department of General Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Jiaxin Hu
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, 445000, Hubei, China
| | - Bing Song
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Ya Hou
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Jinxuan Zhao
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Haiting Chen
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Jun Xie
- The Affiliated Drum Tower Hospital of Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
115
|
Ni X, Gao C, Zhu X, Zhang X, Fang Y, Hao Z. Isobavachalcone induces hepatotoxicity in zebrafish embryos and HepG2 cells via the System Xc --GSH-GPX4 signaling pathway in ferroptosis response. J Appl Toxicol 2024; 44:1139-1152. [PMID: 38581191 DOI: 10.1002/jat.4607] [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: 02/23/2024] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 04/08/2024]
Abstract
Isobavachalcone (IBC) is a flavonoid component of the traditional Chinese medicine Psoraleae Fructus, with a range of pharmacological properties. However, IBC causes some hepatotoxicity, and the mechanism of toxicity is unclear. The purpose of this paper was to investigate the possible mechanism of toxicity of IBC on HepG2 cells and zebrafish embryos. The results showed that exposure to IBC increased zebrafish embryo mortality and decreased hatchability. Meanwhile, IBC induced liver injury and increased expression of ALT and AST activity. Further studies showed that IBC caused the increase of ROS and MDA the decrease of CAT, GSH, and GSH-Px; the increase of Fe2+ content; and the changes of ferroptosis related genes (acsl4, gpx4, and xct) and iron storage related genes (tf, fth, and fpn) in zebrafish embryos. Through in vitro verification, it was found that IBC also caused oxidative stress and increased Fe2+ content in HepG2 cells. IBC caused depolarization of mitochondrial membrane potential (MMP) and reduction of mitochondrial ATP, as well as altered expression of ACSl4, SLC7A11, GPX4, and FTH1 proteins. Treatment of HepG2 cells with ferrostatin-1 could reverse the effect of IBC. Targeting the System Xc--GSH-GPX4 pathway of ferroptosis and preventing oxidative stress damage might offer a theoretical foundation for practical therapy and prevention of IBC-induced hepatotoxicity.
Collapse
Affiliation(s)
- Xuan Ni
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chen Gao
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaolin Zhu
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaosong Zhang
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yizhuo Fang
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhihui Hao
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
116
|
Guan S, Zhang S, Liu M, Guo J, Chen Y, Shen X, Deng X, Lu J. Preventive effects of lactoferrin on acute alcohol-induced liver injury via iron chelation and regulation of iron metabolism. J Dairy Sci 2024; 107:5316-5329. [PMID: 38608952 DOI: 10.3168/jds.2023-24490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/02/2024] [Indexed: 04/14/2024]
Abstract
Lactoferrin is widely found in milk and has the ability to bind iron. Previous studies have reported that lactoferrin was effective in the prevention and treatment of acute alcohol-induced liver injury (AALI). Ferroptosis is a recently discovered cell death and is involved in the development of AALI. However, the potential role of lactoferrin in acute alcohol-induced ferroptosis is still unclear. In this study, we observed that lactoferrin (10, 20, and 40 μg/mL) significantly mitigated alcohol (300 mM)-induced injury in vitro. Additionally, lactoferrin (100 and 200 mg/kg BW) significantly alleviated alcohol (4.8 g/kg BW)-induced injury in vivo. Our results showed that lactoferrin inhibited alcohol-induced upregulation of the ferroptosis marker protein ACSL4 and downregulation of GPX4. Meanwhile, lactoferrin treatment successfully reversed the elevated malondialdehyde (MDA) levels and the reduced glutathione (GSH) levels caused by alcohol treatment. These results may indicate that lactoferrin significantly decreased ferroptosis in vivo and in vitro. Lactoferrin has the potential to chelate iron, and our results showed that lactoferrin (20 μg/mL) significantly reduced iron ions and the expression of the ferritin heavy chain (FTH) under FeCl3 (100 μM) treatment. It was demonstrated that lactoferrin had a significant iron-chelating effect and reduced iron overload caused by FeCl3 in AML12 cells. Next, we examined iron content and the expression of iron metabolism marker proteins transferrin receptor (TFR), divalent metal transporter 1 (DMT1), FTH, and ferroportin (FPN). Our results showed that lactoferrin alleviated iron overload induced by acute alcohol. The expression of TFR and DMT1 was downregulated, and FPN and FTH were upregulated after lactoferrin treatment in vivo and in vitro. Above all, the study suggested that lactoferrin can alleviate AALI by mitigating acute alcohol-induced ferroptosis. Lactoferrin may offer new strategies for the prevention or treatment of AALI.
Collapse
Affiliation(s)
- Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China; State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Shengzhuo Zhang
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Meitong Liu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Jiakang Guo
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Yuelin Chen
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Xue Shen
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China.
| | - Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China.
| |
Collapse
|
117
|
Chen Q, Wang Y, Wang J, Ouyang X, Zhong J, Huang Y, Huang Z, Zheng B, Peng L, Tang X, Li S. Lipotoxicity Induces Cardiomyocyte Ferroptosis via Activating the STING Pathway. Antioxid Redox Signal 2024. [PMID: 39001814 DOI: 10.1089/ars.2023.0510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/15/2024]
Affiliation(s)
- Qian Chen
- Department of Cardiovascular Medicine, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yina Wang
- VIP medical service center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiafu Wang
- Department of Cardiovascular Medicine, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaolan Ouyang
- Department of Cardiovascular Medicine, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junlin Zhong
- Department of Ultrasonography, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yao Huang
- Zhongshan School of Medicine, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuoshan Huang
- Department of Cardiovascular Medicine, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Benrong Zheng
- VIP medical service center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Long Peng
- Department of Cardiovascular Medicine, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xixiang Tang
- VIP medical service center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Suhua Li
- Department of Cardiovascular Medicine, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
118
|
Gao HX, Jiang J, Yang CY, Xu JF, He Q, Hu YW. Zinc finger translocation‑associated protein promotes ferroptosis through the upregulation of ACSL4 expression in vascular endothelial cells. Exp Ther Med 2024; 28:334. [PMID: 39011065 PMCID: PMC11247542 DOI: 10.3892/etm.2024.12623] [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: 11/28/2023] [Accepted: 05/17/2024] [Indexed: 07/17/2024] Open
Abstract
Numerous studies have reported the potential involvement of ferroptosis in the development of atherosclerosis (AS). Acyl-CoA synthetase long chain family member 4 (ACSL4) is an essential component in the promotion of ferroptosis. The present study aimed to investigate the role of ACSL4 and zinc finger translocation-associated protein (ZFTA) in the regulation of endothelial cell ferroptosis in AS. Human umbilical vein endothelial cells (HUVECs) with ACSL4 knockout were generated using CRISPR/Cas9 technology. To assess ferroptosis, malondialdehyde concentration, iron content and reactive oxygen species levels were quantified in the present study. In addition, western blot analysis was conducted to explore the potential mechanisms underlying ACSL4 and ZFTA in the modulation of ferroptosis in HUVECs. The results of the present study demonstrated that the expression levels of ACSL4 and ZFTA were significantly increased in human atherosclerotic plaques. In addition, ACSL4 knockout led to a reduced susceptibility to ferroptosis, while ZFTA contributed to ferroptosis in HUVECs. Results of the present study also demonstrated that ZFTA overexpression upregulated ACSL4 expression in HUVECs, whereas ZFTA knockdown led to decreased ACSL4 expression. Co-transfection experiments demonstrated that the ZTFA overexpression-mediated increase in ferroptosis was reversed following ACSL4 knockdown. Collectively, results of the present study highlighted that ACSL4 mediated the effects of ZFTA on the ferroptosis of HUVECs. Thus, the present study demonstrated the potential role of ACSL4 and ZFTA in the regulation of ferroptosis, and highlighted that ferroptosis-related pathways may act as potential targets in the treatment of AS.
Collapse
Affiliation(s)
- Hui-Xin Gao
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Jun Jiang
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Chun-Yan Yang
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Jin-Fu Xu
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Qing He
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Yan-Wei Hu
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| |
Collapse
|
119
|
Zhang J, Nie C, Zhang Y, Yang L, Du X, Liu L, Chen Y, Yang Q, Zhu X, Li Q. Analysis of mechanism, therapeutic strategies, and potential natural compounds against atherosclerosis by targeting iron overload-induced oxidative stress. Biomed Pharmacother 2024; 177:117112. [PMID: 39018869 DOI: 10.1016/j.biopha.2024.117112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/04/2024] [Accepted: 07/07/2024] [Indexed: 07/19/2024] Open
Abstract
Ferroptosis is a novel form of cell demise characterized primarily by the reduction of trivalent iron to divalent iron, leading to the release of reactive oxygen species (ROS) and consequent induction of intense oxidative stress. In atherosclerosis (AS), highly accumulated lipids are modified by ROS to promote the formation of lipid peroxides, further amplifying cellular oxidative stress damage to influence all stages of atherosclerotic development. Macrophages are regarded as pivotal executors in the progression of AS and the handling of iron, thus targeting macrophage iron metabolism holds significant guiding implications for exploring potential therapeutic strategies against AS. In this comprehensive review, we elucidate the potential interplay among iron overload, inflammation, and lipid dysregulation, summarizing the potential mechanisms underlying the suppression of AS by alleviating iron overload. Furthermore, the application of Traditional Chinese Medicine (TCM) is increasingly widespread. Based on extant research and the pharmacological foundations of active compounds of TCM, we propose alternative therapeutic agents for AS in the context of iron overload, aiming to diversify the therapeutic avenues.
Collapse
Affiliation(s)
- Jing Zhang
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Chunxia Nie
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Yang Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Lina Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Xinke Du
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China.
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China; State key laboratory for quality ensurance and sustainable use ofdao-di herbs, Beijing 100700, China.
| |
Collapse
|
120
|
Oliveira CA, Mercês ÉAB, Portela FS, Malheiro LFL, Silva HBL, De Benedictis LM, De Benedictis JM, Silva CCDE, Santos ACL, Rosa DP, Velozo HS, de Jesus Soares T, de Brito Amaral LS. An integrated view of cisplatin-induced nephrotoxicity, hepatotoxicity, and cardiotoxicity: characteristics, common molecular mechanisms, and current clinical management. Clin Exp Nephrol 2024; 28:711-727. [PMID: 38678166 DOI: 10.1007/s10157-024-02490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/18/2024] [Indexed: 04/29/2024]
Abstract
Cisplatin (CP) is a chemotherapy drug widely prescribed to treat various neoplasms. Although fundamental for the therapeutic action of the drug, its cytotoxic mechanisms trigger adverse effects in several tissues, such as the kidney, liver, and heart, which limit its clinical use. In this sense, studies point to an essential role of damage to nuclear and mitochondrial DNA associated with oxidative stress, inflammation, and apoptosis in the pathophysiology of tissue injuries. Due to the limitation of effective preventive and therapeutic measures against CP-induced toxicity, new strategies with potential cytoprotective effects have been studied. Therefore, this article is timely in reviewing the characteristics and main molecular mechanisms common to renal, hepatic, and cardiac toxicity previously described, in addition to addressing the main validated strategies for the current management of these adverse events in clinical practice. We also handle the main promising antioxidant substances recently presented in the literature to encourage the development of new research that consolidates their potential preventive and therapeutic effects against CP-induced cytotoxicity.
Collapse
Affiliation(s)
- Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Érika Azenathe Barros Mercês
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Lara Fabiana Luz Malheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | | | | | | | | | | | | | - Helloisa Souza Velozo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
| |
Collapse
|
121
|
Zhao R, Chen Y, Liang Y. Bioorthogonal Delivery of Carbon Disulfide in Living Cells. Angew Chem Int Ed Engl 2024; 63:e202400020. [PMID: 38752888 DOI: 10.1002/anie.202400020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Indexed: 06/27/2024]
Abstract
Carbon disulfide (CS2) is an environmental contaminant, which is deadly hazardous to the workers under chronic or acute exposure. However, the toxicity mechanisms of CS2 are still unclear due to the scarcity of biocompatible donors, which can release CS2 in cells. Here we developed the first bioorthogonal CS2 delivery system based on the "click-and-release" reactions between mesoionic 1,3-thiazolium-5-thiolates (TATs) and strained cyclooctyne exo-BCN-OH. We successfully realized intracellular CS2 release and investigated the causes of CS2-induced hepatotoxicity, including oxidative stress, proteotoxic stress and copper-dependent cell death. It is found that CS2 can be copper vehicles bypassing copper transporters after reacting with nucleophiles in cytoplasm, and extra copper supplementation will exacerbate the loss of homeostasis of cells and ultimately cell death. These findings inspired us to explore the anticancer activity of CS2 in combination with copper by introducing a copper chelating group in our CS2 delivery system.
Collapse
Affiliation(s)
- Ruohan Zhao
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yinghan Chen
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yong Liang
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
122
|
He X, Long Q, Zhong Y, Zhang Y, Qian B, Huang S, Chang L, Qi Z, Li L, Wang X, Yang X, Dong Gao W, Ye X, Zhao Q. Short-chain fatty acids regulate erastin-induced cardiomyocyte ferroptosis and ferroptosis-related genes. Front Pharmacol 2024; 15:1409321. [PMID: 39070785 PMCID: PMC11272585 DOI: 10.3389/fphar.2024.1409321] [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: 03/29/2024] [Accepted: 06/07/2024] [Indexed: 07/30/2024] Open
Abstract
Background Ferroptosis has been proven to contribute to the progression of myocardial ischemia/reperfusion (I/R) injury and can be inhibited or promoted by ATF3. Short-chain fatty acids (SCFAs) have shown benefits in various cardiovascular diseases with anti-inflammatory and antioxidant effects. However, the impact of SCFAs on ferroptosis in ischemic-stimulated cardiomyocytes remains unknown. This study aimed to investigate the effect of SCFAs on cardiomyocyte ferroptosis, the expression of ATF3, and its potential upstream regulators. Methods and results The expression of ATF3, ferroptosis pathway geneset (FPG), and geneset of potential regulators for ATF3 (GPRA, predicted by the PROMO database) was explored in the public human myocardial infarction single-cell RNA-seq (sma) dataset. Cardiomyocyte data was extracted from the dataset and re-clustered to explore the FPG, ATF3, and GPRA expression patterns in cardiomyocyte subclusters. A dose-dependent toxic experiment was run to detect the suitable dose for SCFA treatment. The erastin-induced ferroptosis model and hypoxia-reoxygenation (H/R) model (10 h of hypoxia followed by 6 h of reoxygenation) were adopted to assess the effect of SCFAs via the CCK8 assay. Gene expression was examined via RT-PCR and western blot. Ferroptosis markers, including lipid peroxides and Fe2+, were detected using the liperfluo and ferroOrange probes, respectively. In the sma dataset, upregulated ferroptosis pathway genes were mainly found in the infarction-stimulated cardiac cells (border zone and fibrotic zone), particularly the cardiomyocytes and adipocytes. The ATF3 and some of its potential transcription factors (VDR, EGR3, PAX5, and SP1) can be regulated by SCFA. SCFA can attenuate erastin-induced lipid peroxidation in cardiomyocytes. SCFA treatment can also reverse erastin-induced Fe2+ increase but may strengthen the Fe2+ in the H/R model. We also precisely defined a ferroptosis subcluster of cardiomyocytes (CM09) that highly expressed FPG, ATF3, and GPRA. Conclusion The ATF3 and the ferroptosis pathway are elevated in cardiomyocytes of injury-related cardiac regions (border zone, ischemic zone, and fibrotic zone). SCFA can attenuate cardiomyocyte ferroptosis and regulate the expression of ATF3. Our study offers novel insights into the potential targets of SCFAs in the cardiovascular system.
Collapse
Affiliation(s)
- Xiaojun He
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Long
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiming Zhong
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yecen Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bei Qian
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shixing Huang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lan Chang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaoxi Qi
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lihui Li
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xinming Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaomei Yang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaofeng Ye
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
123
|
Matheus J, Alegria MJ, Nunes MC, Raymundo A. Algae-Boosted Chickpea Hummus: Improving Nutrition and Texture with Seaweeds and Microalgae. Foods 2024; 13:2178. [PMID: 39063262 PMCID: PMC11276347 DOI: 10.3390/foods13142178] [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: 05/29/2024] [Revised: 07/04/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
The global food industry faces a critical challenge in ensuring sustainable practices to meet the demands of a growing population while minimizing environmental impact. At the same time, consumer awareness and the demand for quality products drive innovation and inspire positive changes in the food supply chain. Aiming to create a more sustainable and nutrient-rich alternative, this study is summarized by characterizing the physical and chemical characteristics of algae-enriched chickpea hummus: an innovative approach to popular food products. The algae-enriched hummuses were developed with an incorporation (6% w/w) of Gelidium corneum and Fucus vesiculosus seaweeds and Chlorella vulgaris (hetero and autotrophic) microalgae to reveal their technological potential and evaluate the nutritional and rheological characteristics relative to a control hummus (without algae). From a nutritional perspective, the main results indicated that hummus enriched with microalgae showed an increase in protein content and an improved mineral profile. This was particularly notable for the seaweed F. vesiculosus and the autotrophic microalga C. vulgaris, leading to claims of being a "source of" and "rich in" various minerals. Additionally, the antioxidant activity of hummus containing F. vesiculosus and C. vulgaris increased significantly compared to the control. From a rheological perspective, incorporating algae into the humus strengthened its structure. The microalgae further enhanced the dish's elasticity and firmness, thus improving this chickpea-based dish´s overall texture and quality.
Collapse
Affiliation(s)
- José Matheus
- LEAF—Linking Landscape, Environment, Agriculture and Food Research Center, Associated Laboratory TERRA, Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; (J.M.); (M.C.N.)
| | - Maria João Alegria
- SUMOL+COMPAL, Rua Dr. António João Eusébio, 24, 2790-179 Carnaxide, Portugal;
| | - Maria Cristiana Nunes
- LEAF—Linking Landscape, Environment, Agriculture and Food Research Center, Associated Laboratory TERRA, Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; (J.M.); (M.C.N.)
| | - Anabela Raymundo
- LEAF—Linking Landscape, Environment, Agriculture and Food Research Center, Associated Laboratory TERRA, Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; (J.M.); (M.C.N.)
| |
Collapse
|
124
|
Packer M, Anker SD, Butler J, Cleland JG, Kalra PR, Mentz RJ, Ponikowski P, Talha KM. Redefining Iron Deficiency in Patients With Chronic Heart Failure. Circulation 2024; 150:151-161. [PMID: 38733252 PMCID: PMC11224570 DOI: 10.1161/circulationaha.124.068883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024]
Abstract
A serum ferritin level <15 to 20 μg/L historically identified patients who had absent bone marrow iron stores, but serum ferritin levels are distorted by the systemic inflammatory states seen in patients with chronic kidney disease or heart failure. As a result, nearly 25 years ago, the diagnostic ferritin threshold was increased 5- to 20-fold in patients with chronic kidney disease (ie, iron deficiency was identified if the serum ferritin level was <100 μg/L, regardless of transferrin saturation [TSAT], or 100 to 299 μg/L if TSAT was <20%). This guidance was motivated not by the findings of studies of total body or tissue iron depletion, but by a desire to encourage the use of iron supplements to potentiate the response to erythropoiesis-stimulating agents in patients with renal anemia. However, in patients with heart failure, this definition does not reliably identify patients with an absolute or functional iron-deficiency state, and it includes individuals with TSATs (≥20%) and serum ferritin levels in the normal range (20-100 mg/L) who are not iron deficient, have an excellent prognosis, and do not respond favorably to iron therapy. Furthermore, serum ferritin levels may be distorted by the use of both neprilysin and sodium-glucose cotransporter 2 inhibitors, both of which may act to mobilize endogenous iron stores. The most evidence-based and trial-tested definition of iron deficiency is the presence of hypoferremia, as reflected by as a TSAT <20%. These hypoferremic patients are generally iron deficient on bone marrow examination, and after intravenous iron therapy, they exhibit an improvement in exercise tolerance and functional capacity (when meaningfully impaired) and show the most marked reduction (ie, 20%-30%) in the risk of cardiovascular death or total heart failure hospitalizations. Therefore, we propose that the current ferritin-driven definition of iron deficiency in heart failure should be abandoned and that a definition based on hypoferremia (TSAT <20%) should be adopted.
Collapse
Affiliation(s)
- Milton Packer
- Baylor University Medical Center (M.P.), Dallas, TX
- Imperial College, London, UK (M.P.)
| | - Stefan D. Anker
- Department of Cardiology, German Heart Center Charité, Institute of Health Center for Regenerative Therapies, German Centre for Cardiovascular Research, partner site Berlin, Charité Universitätsmedizin, Berlin, Germany (S.D.A.)
| | - Javed Butler
- Baylor Scott and White Research Institute (J.B.), Dallas, TX
- Baylor University Medical Center (M.P.), Dallas, TX
- University of Mississippi Medical Center, Jackson (J.B., K.M.T.)
| | - John G.F. Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health (J.G.F.C.), University of Glasgow, UK
| | - Paul R. Kalra
- College of Medical, Veterinary & Life Sciences (P.R.K.), University of Glasgow, UK
- Department of Cardiology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK (P.R.K.)
- Faculty of Science and Health, University of Portsmouth, UK (P.R.K.)
| | - Robert J. Mentz
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.M.)
- Duke Clinical Research Institute, Durham, NC (R.J.M.)
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Poland (P.P.)
- Institute of Heart Diseases, University Hospital, Wroclaw, Poland (P.P.)
| | - Khawaja M. Talha
- University of Mississippi Medical Center, Jackson (J.B., K.M.T.)
| |
Collapse
|
125
|
Fujii J, Imai H. Oxidative Metabolism as a Cause of Lipid Peroxidation in the Execution of Ferroptosis. Int J Mol Sci 2024; 25:7544. [PMID: 39062787 PMCID: PMC11276677 DOI: 10.3390/ijms25147544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Ferroptosis is a type of nonapoptotic cell death that is characteristically caused by phospholipid peroxidation promoted by radical reactions involving iron. Researchers have identified many of the protein factors that are encoded by genes that promote ferroptosis. Glutathione peroxidase 4 (GPX4) is a key enzyme that protects phospholipids from peroxidation and suppresses ferroptosis in a glutathione-dependent manner. Thus, the dysregulation of genes involved in cysteine and/or glutathione metabolism is closely associated with ferroptosis. From the perspective of cell dynamics, actively proliferating cells are more prone to ferroptosis than quiescent cells, which suggests that radical species generated during oxygen-involved metabolism are responsible for lipid peroxidation. Herein, we discuss the initial events involved in ferroptosis that dominantly occur in the process of energy metabolism, in association with cysteine deficiency. Accordingly, dysregulation of the tricarboxylic acid cycle coupled with the respiratory chain in mitochondria are the main subjects here, and this suggests that mitochondria are the likely source of both radical electrons and free iron. Since not only carbohydrates, but also amino acids, especially glutamate, are major substrates for central metabolism, dealing with nitrogen derived from amino groups also contributes to lipid peroxidation and is a subject of this discussion.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Hirotaka Imai
- Laboratory of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, Tokyo 108-8641, Japan
- Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo 108-8641, Japan
| |
Collapse
|
126
|
Tan M, Li W, He H, Wang J, Chen Y, Guo Y, Lin T, Ke F. Targeted mitochondrial fluorescence probe with large stokes shift for detecting viscosity changes in vivo and in ferroptosis process. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 315:124246. [PMID: 38593540 DOI: 10.1016/j.saa.2024.124246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/21/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
We created four fluorescent sensors in our work to determine the viscosity of mitochondria. Following screening, the probe Mito-3 was chosen because in contrast to the other three probes, it had a greater fluorescence enhancement, large Stokes shift (113 nm) and had a particular response to viscosity that was unaffected by polarity or biological species. As the viscosity increased from PBS to 90 % glycerol, the fluorescence intensity of probe at 586 nm increased 17-fold. Mito-3 has strong biocompatibility and is able to track changes in cell viscosity in response to nystatin and monensin stimulation. Furthermore, the probe has been successfully applied to detect changes in viscosity caused by nystatin and monensin in zebrafish. Above all, the probe can be applied to the increase in mitochondrial viscosity that accompanies the ferroptosis process. Mito-3 has the potential to help further study the relationship between viscosity and ferroptosis.
Collapse
Affiliation(s)
- Meixia Tan
- School of Pharmacy, Institute of Materia Medica, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350004, China
| | - Wei Li
- School of Pharmacy, Institute of Materia Medica, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350004, China
| | - Hongxing He
- Fujian Medical University Laboratory Animal Center, Fujian Medical University, Fuzhou 350004, China
| | - Jin Wang
- School of Pharmacy, Institute of Materia Medica, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350004, China
| | - Yan Chen
- School of Pharmacy, Institute of Materia Medica, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350004, China
| | - Yuelin Guo
- School of Pharmacy, Institute of Materia Medica, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350004, China
| | - Tiansheng Lin
- Department of Nuclear Medicine, Fujian Medical University Union Hospital, Fuzhou 350004, China.
| | - Fang Ke
- School of Pharmacy, Institute of Materia Medica, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350004, China.
| |
Collapse
|
127
|
Zeng L, Yang K, Yu G, Hao W, Zhu X, Ge A, Chen J, Sun L. Advances in research on immunocyte iron metabolism, ferroptosis, and their regulatory roles in autoimmune and autoinflammatory diseases. Cell Death Dis 2024; 15:481. [PMID: 38965216 PMCID: PMC11224426 DOI: 10.1038/s41419-024-06807-2] [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: 02/10/2024] [Revised: 05/26/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
Autoimmune diseases commonly affect various systems, but their etiology and pathogenesis remain unclear. Currently, increasing research has highlighted the role of ferroptosis in immune regulation, with immune cells being a crucial component of the body's immune system. This review provides an overview and discusses the relationship between ferroptosis, programmed cell death in immune cells, and autoimmune diseases. Additionally, it summarizes the role of various key targets of ferroptosis, such as GPX4 and TFR, in immune cell immune responses. Furthermore, the release of multiple molecules, including damage-associated molecular patterns (DAMPs), following cell death by ferroptosis, is examined, as these molecules further influence the differentiation and function of immune cells, thereby affecting the occurrence and progression of autoimmune diseases. Moreover, immune cells secrete immune factors or their metabolites, which also impact the occurrence of ferroptosis in target organs and tissues involved in autoimmune diseases. Iron chelators, chloroquine and its derivatives, antioxidants, chloroquine derivatives, and calreticulin have been demonstrated to be effective in animal studies for certain autoimmune diseases, exerting anti-inflammatory and immunomodulatory effects. Finally, a brief summary and future perspectives on the research of autoimmune diseases are provided, aiming to guide disease treatment strategies.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China.
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China.
| | - Ganpeng Yu
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Wensa Hao
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junpeng Chen
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China.
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, USA.
- College of Mechanical Engineering, Hunan University of Science and Technology, Xiangtan, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
128
|
Maimaiti Y, Su T, Zhang Z, Ma L, Zhang Y, Xu H. NOX4-mediated astrocyte ferroptosis in Alzheimer's disease. Cell Biosci 2024; 14:88. [PMID: 38956702 PMCID: PMC11218381 DOI: 10.1186/s13578-024-01266-w] [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: 01/11/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
This study investigates NADPH oxidase 4 (NOX4) involvement in iron-mediated astrocyte cell death in Alzheimer's Disease (AD) using single-cell sequencing data and transcriptomes. We analyzed AD single-cell RNA sequencing data, identified astrocyte marker genes, and explored biological processes in astrocytes. We integrated AD-related chip data with ferroptosis-related genes, highlighting NOX4. We validated NOX4's role in ferroptosis and AD in vitro and in vivo. Astrocyte marker genes were enriched in AD, emphasizing their role. NOX4 emerged as a crucial player in astrocytic ferroptosis in AD. Silencing NOX4 mitigated ferroptosis, improved cognition, reduced Aβ and p-Tau levels, and alleviated mitochondrial abnormalities. NOX4 promotes astrocytic ferroptosis, underscoring its significance in AD progression.
Collapse
Affiliation(s)
- Yasenjiang Maimaiti
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No.91 Tianchi Road, Urumqi, Xinjiang, China.
| | - Ting Su
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No.91 Tianchi Road, Urumqi, Xinjiang, China
| | - Zhanying Zhang
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No.91 Tianchi Road, Urumqi, Xinjiang, China
| | - Lingling Ma
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No.91 Tianchi Road, Urumqi, Xinjiang, China
| | - Yuan Zhang
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No.91 Tianchi Road, Urumqi, Xinjiang, China
| | - Hong Xu
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No.91 Tianchi Road, Urumqi, Xinjiang, China.
| |
Collapse
|
129
|
Gvozdenko A, Blinov A, Golik A, Rekhman Z, Nagdalian A, Filippov D, Askerova A, Bocharov N, Kastarnova E, Hassan FA, AL-Farga A, Shariati MA. Harnessing the Power of a Novel Triple Chelate Complex in Fermented Probiotic Dairy Products: A Promising Solution for Combating Iron Deficiency Anemia. ACS OMEGA 2024; 9:28594-28610. [PMID: 38973905 PMCID: PMC11223220 DOI: 10.1021/acsomega.4c02664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024]
Abstract
This study discovered and examined novel triple chelate complexes involving iron, ascorbic acid, and essential amino acids (AsA-Fe-AmA triple chelate complexes) for the first time. The mechanism of complex formation was studied using FTIR spectroscopy and quantum chemical modeling. The produced complexes were shown to be suitable for fortifying food items with a pH of 3-7 that have not been exposed to heat treatment at temperatures over 75 °C for more than 15 min. Thus, it can be said that the concentration for milk fortification should be 0.005 mol/L or less. In vivo experiments in rats models revealed that the synthesized complexes increased serum iron levels after a single application to reference values within 24 h of oral administration. The iron level increased by 14.0 mmol/L at 2 mL dose of the complex. This fact makes it possible to consider the use of developed complexes and developed fermented dairy products for the prevention of iron deficiency and iron deficiency anemia. Research on the effect of discovered compounds on the physicochemical and organoleptic qualities of milk was conducted. Furthermore, iron ascorbate threoninate, iron ascorbate methioninate, iron ascorbate lysinate, and iron ascorbate tryptophanate all had a beneficial effect on Lacticaseibacillus rhamnosus at concentrations as low as 0.0005 mol/L, which is significant for milk fermentation. A study of fermented milk products revealed that the most effective AsA-Fe-AmA triple chelate complex is iron ascorbate lysinate, which might be further investigated as a viable molecule for dietary fortification in iron deficiency anemia. It was found that fortified fermented milk products had a titratable acidity of 67 ± 1°T, pH of 4.38 ± 0.05, and a viscosity of 2018 ± 142 Pa·s.
Collapse
Affiliation(s)
- Alexey Gvozdenko
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Andrey Blinov
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Alexey Golik
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Zafar Rekhman
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Andrey Nagdalian
- Laboratory
of Food and Industrial Biotechnology, North-Caucasus
Federal University, Stavropol 355017, Russia
| | - Dionis Filippov
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Alina Askerova
- Laboratory
of Food and Industrial Biotechnology, North-Caucasus
Federal University, Stavropol 355017, Russia
| | - Nikita Bocharov
- Physical
and Technical Faculty, North-Caucasus Federal
University, Stavropol 355017, Russia
| | - Elena Kastarnova
- Eterinary
Faculty, Stavropol Sate Agrarian University, Zootechnicheskiy Street 9, Stavropol 355017, Russia
| | - Faten Abdo Hassan
- Faculty
of Science, Department of Microbiology, Taiz University, Taiz 9674, Yemen
| | - Ammar AL-Farga
- Department
of Biochemistry, College of Science, University
of Jeddah, Jeddah 21577, Saudi Arabia
| | - Mohammad Ali Shariati
- Scientific
Department, Semey Branch of the Kazakh Research
Institute of Processing and Food Industry, Gagarin Avenue 238G, Almaty 050060, Kazakhstan
| |
Collapse
|
130
|
Lv Q, Lin J, Huang H, Ma B, Li W, Chen J, Wang M, Wang X, Fu G, Xiao Y. Nanosponge for Iron Chelation and Efflux: A Ferroptosis-Inhibiting Approach for Myocardial Infarction Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305895. [PMID: 38671590 PMCID: PMC11220697 DOI: 10.1002/advs.202305895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 04/11/2024] [Indexed: 04/28/2024]
Abstract
Myocardial infarction (MI), a consequence of coronary artery occlusion, triggers the degradation of ferritin, resulting in elevated levels of free iron in the heart and thereby inducing ferroptosis. Targeting myocardial ferroptosis through the chelation of excess iron has therapeutic potential for MI treatment. However, iron chelation in post ischemic injury areas using conventional iron-specific chelators is hindered by ineffective myocardial intracellular chelation, rapid clearance, and high systemic toxicity. A chitosan-desferrioxamine nanosponge (CDNS) is designed by co-crosslinking chitosan and deferoxamine through noncovalent gelation to address these challenges. This architecture facilitates direct iron chelation regardless of deferoxamine (DFO) release due to its sponge-like porous hydrogel structure. Upon cellular internalization, CDNS can effectively chelate cellular iron and facilitate the efflux of captured iron, thereby inhibiting ferroptosis and associated oxidative stress and lipid peroxidation. In MI mouse models, myocardial injection of CDNS promotes sustainable retention and the suppression of ferroptosis in the infarcted heart. This intervention improves cardiac function and alleviates adverse cardiac remodeling post-MI, leading to decreased oxidative stress and the promotion of angiogenesis due to ferroptosis inhibition by CDNS in the infarcted heart. This study reveals a nanosponge-based nanomedicine targeting myocardial ferroptosis with efficient iron chelation and efflux, offering a promising MI treatment.
Collapse
Affiliation(s)
- Qingbo Lv
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Jun Lin
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
- Department of Cardiovascular SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - He Huang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Boxuan Ma
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Wujiao Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Jiawen Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Xiaoyu Wang
- Qiushi Academy for Advanced StudiesZhejiang UniversityHangzhou310058China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Yun Xiao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| |
Collapse
|
131
|
Li Z, Zhang Y, Ji M, Wu C, Zhang Y, Ji S. Targeting ferroptosis in neuroimmune and neurodegenerative disorders for the development of novel therapeutics. Biomed Pharmacother 2024; 176:116777. [PMID: 38795640 DOI: 10.1016/j.biopha.2024.116777] [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: 03/21/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
Neuroimmune and neurodegenerative ailments impose a substantial societal burden. Neuroimmune disorders involve the intricate regulatory interactions between the immune system and the central nervous system. Prominent examples of neuroimmune disorders encompass multiple sclerosis and neuromyelitis optica. Neurodegenerative diseases result from neuronal degeneration or demyelination in the brain or spinal cord, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. The precise underlying pathogenesis of these conditions remains incompletely understood. Ferroptosis, a programmed form of cell death characterised by lipid peroxidation and iron overload, plays a pivotal role in neuroimmune and neurodegenerative diseases. In this review, we provide a detailed overview of ferroptosis, its mechanisms, pathways, and regulation during the progression of neuroimmune and neurodegenerative diseases. Furthermore, we summarise the impact of ferroptosis on neuroimmune-related cells (T cells, B cells, neutrophils, and macrophages) and neural cells (glial cells and neurons). Finally, we explore the potential therapeutic implications of ferroptosis inhibitors in diverse neuroimmune and neurodegenerative diseases.
Collapse
Affiliation(s)
- Zihao Li
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China
| | - Ye Zhang
- Department of Forensic Medicine, Shantou University Medical College (SUMC), Shantou, Guangdong, China
| | - Meiling Ji
- Department of Emergency, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 210002, China
| | - Chenglong Wu
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China
| | - Yanxing Zhang
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China.
| | - Senlin Ji
- Department of Neurology of Nanjing Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Translational Medicine Institute of Brain Disorders, Nanjing University, Nanjing, Jiangsu 210008, China.
| |
Collapse
|
132
|
He Y, Wang X, Chen S, Luo H, Huo B, Guo X, Li R, Chen Y, Yi X, Wei X, Jiang DS. SP2509 functions as a novel ferroptosis inhibitor by reducing intracellular iron level in vascular smooth muscle cells. Free Radic Biol Med 2024; 219:49-63. [PMID: 38608823 DOI: 10.1016/j.freeradbiomed.2024.04.220] [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: 10/30/2023] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Previous studies have shown that ferroptosis of vascular smooth muscle cells (VSMCs) is involved in the development of aortic dissection (AD) and that histone methylation regulates this process. SP2509 acts as a specific inhibitor of lysine-specific demethylase 1 (LSD1), which governs a variety of biological processes. However, the effect of SP2509 on VSMC ferroptosis and AD remains to be elucidated. This aim of this study was to investigate the role and underlying mechanism of SP2509-mediated histone methylation on VSMC ferroptosis. Here, a mouse model of AD was established, and significantly reduced levels of H3K4me1 and H3K4me2 (target of SP2509) were found in the aortas of AD mice. In VSMCs, SP2509 treatment led to a dose-dependent increase in H3K4me2 levels. Furthermore, we found that SP2509 provided equivalent protection to ferrostatin-1 against VSMC ferroptosis, as evidenced by increased cell viability, decreased cell death and lipid peroxidation. RNA-sequencing analysis and subsequent experiments revealed that SP2509 counteracted cystine deficiency-induced response to inflammation and oxidative stress. More importantly, we demonstrated that SP2509 inhibited the expression of TFR and ferritin to reduce intracellular iron levels, thereby effectively blocking the process of ferroptosis. Therefore, our findings indicate that SP2509 protects VSMCs from multiple stimulus-induced ferroptosis by reducing intracellular iron levels, thereby preventing lipid peroxidation and cell death. These findings suggest that SP2509 may be a promising drug to alleviate AD by reducing iron deposition and VSMC ferroptosis.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Mice
- Iron/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Oxidative Stress/drug effects
- Humans
- Disease Models, Animal
- Lipid Peroxidation/drug effects
- Phenylenediamines/pharmacology
- Male
- Cell Survival/drug effects
- Histones/metabolism
- Histones/genetics
- Histone Demethylases/metabolism
- Histone Demethylases/genetics
- Mice, Inbred C57BL
- Cyclohexylamines
Collapse
Affiliation(s)
- Yi He
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xingbo Wang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Siqi Chen
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hanshen Luo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bo Huo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xian Guo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rui Li
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yue Chen
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
133
|
Zhang M, Guo M, Gao Y, Wu C, Pan X, Huang Z. Mechanisms and therapeutic targets of ferroptosis: Implications for nanomedicine design. J Pharm Anal 2024; 14:100960. [PMID: 39135963 PMCID: PMC11318476 DOI: 10.1016/j.jpha.2024.03.001] [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: 10/20/2023] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 08/15/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death and differs considerably from the well-known forms of cell death in terms of cell morphology, genetics, and biochemistry. The three primary pathways for cell ferroptosis are system Xc-/glutathione peroxidase 4 (GPX4), lipid metabolism, and ferric metabolism. Since the discovery of ferroptosis, mounting evidence has revealed its critical regulatory role in several diseases, especially as a novel potential target for cancer therapy, thereby attracting increasing attention in the fields of tumor biology and anti-tumor therapy. Accordingly, broad prospects exist for identifying ferroptosis as a potential therapeutic target. In this review, we aimed to systematically summarize the activation and defense mechanisms of ferroptosis, highlight the therapeutic targets, and discuss the design of nanomedicines for ferroptosis regulation. In addition, we opted to present the advantages and disadvantages of current ferroptosis research and provide an optimistic vision of future directions in related fields. Overall, we aim to provide new ideas for further ferroptosis research and inspire new strategies for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Meihong Zhang
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| | - Mengqin Guo
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| | - Yue Gao
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| | - Chuanbin Wu
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| | - Xin Pan
- College of Pharmacy, University of Sun Yat-sen, Guangzhou, 510275, China
| | - Zhengwei Huang
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| |
Collapse
|
134
|
Chen J, Feng Q, Qiao Y, Pan S, Liang L, Liu Y, Zhang X, Liu D, Liu Z, Liu Z. ACSF2 and lysine lactylation contribute to renal tubule injury in diabetes. Diabetologia 2024; 67:1429-1443. [PMID: 38676722 DOI: 10.1007/s00125-024-06156-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 04/29/2024]
Abstract
AIMS Lactate accumulation is reported to be a biomarker for diabetic nephropathy progression. Lactate drives lysine lactylation, a newly discovered post-translational modification that is involved in the pathogenesis of cancers and metabolic and inflammatory disease. Here, we aimed to determine whether lysine lactylation is involved in the pathogenesis of diabetic nephropathy. METHODS Renal biopsy samples from individuals with diabetic nephropathy (n=22) and control samples from individuals without diabetes and kidney disease (n=9) were obtained from the First Affiliated Hospital of Zhengzhou University for immunohistochemical staining. In addition, we carried out global lactylome profiling of kidney tissues from db/m and db/db mice using LC-MS/MS. Furthermore, we assessed the role of lysine lactylation and acyl-CoA synthetase family member 2 (ACSF2) in mitochondrial function in human proximal tubular epithelial cells (HK-2). RESULTS The expression level of lysine lactylation was significantly increased in the kidneys of individuals with diabetes as well as in kidneys from db/db mice. Integrative lactylome analysis of the kidneys of db/db and db/m mice identified 165 upregulated proteins and 17 downregulated proteins, with an increase in 356 lysine lactylation sites and a decrease in 22 lysine lactylation sites decreased. Subcellular localisation analysis revealed that most lactylated proteins were found in the mitochondria (115 proteins, 269 sites). We further found that lactylation of the K182 site in ACSF2 contributes to mitochondrial dysfunction. Finally, the expression of ACSF2 was notably increased in the kidneys of db/db mice and individuals with diabetic nephropathy. CONCLUSIONS Our study strongly suggests that lysine lactylation and ACSF2 are mediators of mitochondrial dysfunction and may contribute to the progression of diabetic nephropathy. DATA AVAILABILITY The LC-MS/MS proteomics data have been deposited in the ProteomeXchange Consortium database ( https://proteomecentral.proteomexchange.org ) via the iProX partner repository with the dataset identifier PXD050070.
Collapse
Affiliation(s)
- Jingfang Chen
- Traditional Chinese Medicine Integrated Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Qi Feng
- Traditional Chinese Medicine Integrated Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Yingjin Qiao
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shaokang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Lulu Liang
- Traditional Chinese Medicine Integrated Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Yong Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Xiaonan Zhang
- Traditional Chinese Medicine Integrated Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dongwei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Zhangsuo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.
| |
Collapse
|
135
|
Wang Z, Xu B, Tan Y, Fan T. Diagnostic value and method of soluble transferrin receptor for suspected coronary artery disease: a case-control study. Cardiovasc Diagn Ther 2024; 14:402-418. [PMID: 38975011 PMCID: PMC11223933 DOI: 10.21037/cdt-23-450] [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: 11/22/2023] [Accepted: 04/03/2024] [Indexed: 07/09/2024]
Abstract
Background Many studies have pointed out that iron overload in the body is a risk factor for coronary atherosclerosis (AS), while there are also studies that show that iron deficiency is associated with coronary AS. There is still no consensus on how iron metabolism affects coronary artery disease (CAD). This study aimed to analyze the relationship between iron metabolism indexes and CAD, investigate the diagnostic value of soluble transferrin receptor (sTfR) in suspected CAD, and establish a diagnostic model. Methods This was a retrospective study. A total of 268 people with CAD-like symptoms who underwent coronary angiography in the Department of Cardiovascular Medicine, The Second Affiliated Hospital of Anhui Medical University from September 2022 to May 2023 without other chronic diseases or related medication history were included in the study and formed a continuous series including 188 CAD patients and 80 control subjects. Each iron metabolism index was divided into a grade variable according to tertile. The comparison of CAD morbidity between the tertiles and nonlinear correlation test was conducted to investigate the relationship between iron metabolism indexes and CAD risk. We used restricted cubic spline (RCS) to plot the relationship curve between sTfR and CAD risk and to determine the sTfR value corresponding to the minimal odds, according to which we divided the total sample into the "sTfR low level" subgroup and the "sTfR high level" subgroup. Logistic regression analyses were used to establish diagnostic models in both subgroups. The diagnostic efficiency of the indexes and models was compared by receiver operating characteristic (ROC) analysis. Results There is a "J" shape correlation between sTfR and CAD risk. Age/sTfR ratio [area under the curve (AUC) =0.690, 95% confidence interval (CI): 0.598-0.782, specificity 0.488 and sensitivity 0.842] has the best diagnostic efficiency in the "sTfR low level" subgroup. The diagnostic efficiency of sTfR (AUC =0.701, 95% CI: 0.598-0.803, specificity 0.541 and sensitivity 0.797) in the "sTfR high level" subgroup was higher than that of cardiac troponin I (cTnI) (AUC =0.674, 95% CI: 0.564-0.784, specificity 0.719 and sensitivity 0.653). The specific diagnostic methods were as follows: (I) When sTfR ≤1.087 mg/L, calculate the age/sTfR ratio, which indicates the diagnosis of CAD when the result is >58.595; (II) We can directly make a preliminary clinical diagnosis of CAD when sTfR >1.205 mg/L. Except for the above 2 cases, we can initially rule out a diagnosis of CAD. Conclusions The iron metabolism index sTfR correlates with CAD morbidity in a "J" shape. With superior diagnostic efficacy than cTnI, sTfR can assist in diagnosing CAD in patients with CAD-like symptoms. In addition, sTfR can provide guidance for the management of body iron levels in CAD patients.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Banglong Xu
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yan Tan
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tingting Fan
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
136
|
Guo WY, Wang WH, Xu PY, Kankala RK, Chen AZ. Decellularised extracellular matrix-based injectable hydrogels for tissue engineering applications. BIOMATERIALS TRANSLATIONAL 2024; 5:114-128. [PMID: 39351160 PMCID: PMC11438603 DOI: 10.12336/biomatertransl.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 10/04/2024]
Abstract
Decellularised extracellular matrix (dECM) is a biomaterial derived from natural tissues that has attracted considerable attention from tissue engineering researchers due to its exceptional biocompatibility and malleability attributes. These advantageous properties often facilitate natural cell infiltration and tissue reconstruction for regenerative medicine. Due to their excellent fluidity, the injectable hydrogels can be administered in a liquid state and subsequently formed into a gel state in vivo, stabilising the target area and serving in a variety of ways, such as support, repair, and drug release functions. Thus, dECM-based injectable hydrogels have broad prospects for application in complex organ structures and various tissue injury models. This review focuses on exploring research advances in dECM-based injectable hydrogels, primarily focusing on the applications and prospects of dECM hydrogels in tissue engineering. Initially, the recent developments of the dECM-based injectable hydrogels are explained, summarising the different preparation methods with the evaluation of injectable hydrogel properties. Furthermore, some specific examples of the applicability of dECM-based injectable hydrogels are presented. Finally, we summarise the article with interesting prospects and challenges of dECM-based injectable hydrogels, providing insights into the development of these composites in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Wan-Ying Guo
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
| | - Wei-Huang Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
| | - Pei-Yao Xu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian Province, China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian Province, China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian Province, China
| |
Collapse
|
137
|
Camilli M, Viscovo M, Maggio L, Bonanni A, Torre I, Pellegrino C, Lamendola P, Tinti L, Teofili L, Hohaus S, Lanza GA, Ferdinandy P, Varga Z, Crea F, Lombardo A, Minotti G. Sodium-glucose cotransporter 2 inhibitors and the cancer patient: from diabetes to cardioprotection and beyond. Basic Res Cardiol 2024:10.1007/s00395-024-01059-9. [PMID: 38935171 DOI: 10.1007/s00395-024-01059-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/18/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a new drug class initially designed and approved for treatment of diabetes mellitus, have been shown to exert pleiotropic metabolic and direct cardioprotective and nephroprotective effects that extend beyond their glucose-lowering action. These properties prompted their use in two frequently intertwined conditions, heart failure and chronic kidney disease. Their unique mechanism of action makes SGLT2i an attractive option also to lower the rate of cardiac events and improve overall survival of oncological patients with preexisting cardiovascular risk and/or candidate to receive cardiotoxic therapies. This review will cover biological foundations and clinical evidence for SGLT2i modulating myocardial function and metabolism, with a focus on their possible use as cardioprotective agents in the cardio-oncology settings. Furthermore, we will explore recently emerged SGLT2i effects on hematopoiesis and immune system, carrying the potential of attenuating tumor growth and chemotherapy-induced cytopenias.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy.
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy.
| | - Marcello Viscovo
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Maggio
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Alice Bonanni
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Ilaria Torre
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Claudio Pellegrino
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Priscilla Lamendola
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Lorenzo Tinti
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Luciana Teofili
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Stefan Hohaus
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gaetano Antonio Lanza
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltan Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Center of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Rome, Italy
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | | |
Collapse
|
138
|
Li C, Chen H. Role of arachidonic acid in ischemic heart disease under different comorbidities: risk or protection? Cardiovasc Diabetol 2024; 23:218. [PMID: 38915092 PMCID: PMC11197324 DOI: 10.1186/s12933-024-02277-0] [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: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 06/26/2024] Open
Abstract
In a translational study involving animal models and human subjects, Lv et al. demonstrate that arachidonic acid (AA) exhibits cardioprotective effects in diabetic myocardial ischemia, suggesting a departure from its known role in promoting ferroptosis-a form of cell death characterized by iron-dependent lipid peroxidation. However, the study does not address how underlying diabetic conditions might influence the metabolic pathways of AA, which are critical for fully understanding its impact on heart disease. Diabetes can significantly alter lipid metabolism, which in turn might affect the enzymatic processes involved in AA's metabolism, leading to different outcomes in the disease process. Further examination of the role of diabetes in modulating AA's effects could enhance the understanding of its protective mechanism in ischemic conditions. This could also lead to more targeted and effective therapeutic strategies for managing myocardial ischemia in diabetic patients, such as optimizing AA levels to prevent heart damage while avoiding exacerbating factors like ferroptosis.
Collapse
Affiliation(s)
- Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Huijun Chen
- The second affiliated hospital, Heilongjiang University of Chinese Medicine, Harbin, 150001, China.
| |
Collapse
|
139
|
Yoshikawa N, Hirata N, Kurone Y, Shimoeda S. Red ginseng prevents doxorubicin-induced cardiomyopathy by inhibiting cell death via activating the Nrf2 pathway. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:39. [PMID: 38909271 PMCID: PMC11193215 DOI: 10.1186/s40959-024-00242-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Doxorubicin (DXR) is an effective chemotherapeutic agent. DOX-induced cardiomyopathy (DICM), a major limitation of DXR, is a complication with limited treatment options. We previously reported that Red Ginseng (steamed and dried the root of Panax Ginseng cultivated for over six years; RGin) is beneficial for the treatment of DICM. However, the mechanism underlying the action of RGin remains unclear. In this study, we investigated the mechanism of action underlying the efficacy of RGin in the treatment of DICM. METHODS Four-week-old DBA/2 mice were divided into: vehicle, DXR, RGin, and DXR + RGin (n = 10/group). Mice were treated with DXR (4 mg/kg, once a week, accumulated 20 mg/kg, i.p.) or RGin (0.5 g/kg, three times a week, i.p.). To evaluate efficacy, the survival rate and left ventricular ejection fraction (LVEF) were measured as a measure of cardiac function, and cardiomyocytes were subjected to Masson trichrome staining. To investigate the mechanism of action, western blotting was performed to evaluate the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase 1, transferrin receptor (TfR), and other related proteins. Data were analyzed using the Easy R software. Between-group comparisons were performed using one-way analysis of variance and analyzed using a post-hoc Tukey test. Survival rates were estimated using the Kaplan-Meier method and compared using the log-rank test. P < 0.05 was considered statistically significant in all analyses. RESULTS RGin treatment prolongs survival and protects against reduced LVEF. In the DXR group, Nrf2 was not activated and cell death was accelerated. Furthermore, there was an increase in the TfR levels, suggesting abnormal iron metabolism. However, the DXR + RGin group showed activation of the Nrf2 pathway and suppression of myocardial cell death. Furthermore, there was no increase in TfR expression, suggesting that there were no abnormalities in iron metabolism. Therefore, the mechanism of action of RGin in DICM involves an increase in antioxidant activity and inhibition of cell death through activation of the Nrf2 pathway. CONCLUSION RGin is a useful therapeutic candidate for DICM. Its efficacy is supported by the activation of the Nrf2 pathway, which enhances antioxidant activity and inhibits cell death.
Collapse
Affiliation(s)
- Naoki Yoshikawa
- Department of Pharmaceutical Health Care and Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Naoto Hirata
- Department of Pharmaceutical Health Care and Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Yuichiro Kurone
- Department of Pharmaceutical Health Care and Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Sadahiko Shimoeda
- Department of Pharmaceutical Health Care and Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| |
Collapse
|
140
|
Luo Y, Bai XY, Zhang L, Hu QQ, Zhang N, Cheng JZ, Hou MZ, Liu XL. Ferroptosis in Cancer Therapy: Mechanisms, Small Molecule Inducers, and Novel Approaches. Drug Des Devel Ther 2024; 18:2485-2529. [PMID: 38919962 PMCID: PMC11198730 DOI: 10.2147/dddt.s472178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Ferroptosis, a unique form of programmed cell death, is initiated by an excess of iron accumulation and lipid peroxidation-induced damage. There is a growing body of evidence indicating that ferroptosis plays a critical role in the advancement of tumors. The increased metabolic activity and higher iron levels in tumor cells make them particularly vulnerable to ferroptosis. As a result, the targeted induction of ferroptosis is becoming an increasingly promising approach for cancer treatment. This review offers an overview of the regulatory mechanisms of ferroptosis, delves into the mechanism of action of traditional small molecule ferroptosis inducers and their effects on various tumors. In addition, the latest progress in inducing ferroptosis using new means such as proteolysis-targeting chimeras (PROTACs), photodynamic therapy (PDT), sonodynamic therapy (SDT) and nanomaterials is summarized. Finally, this review discusses the challenges and opportunities in the development of ferroptosis-inducing agents, focusing on discovering new targets, improving selectivity, and reducing toxic and side effects.
Collapse
Affiliation(s)
- YiLin Luo
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Xin Yue Bai
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Lei Zhang
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Qian Qian Hu
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Ning Zhang
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Jun Zhi Cheng
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Ming Zheng Hou
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Xiao Long Liu
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| |
Collapse
|
141
|
Zhou J, Shi W, Wu D, Wang S, Wang X, Min J, Wang F. Mendelian Randomization Analysis of Systemic Iron Status and Risk of Different Types of Kidney Disease. Nutrients 2024; 16:1978. [PMID: 38999730 PMCID: PMC11243746 DOI: 10.3390/nu16131978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/08/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
With rapid increases in incidence, diverse subtypes, and complicated etiologies, kidney disease remains a global public health problem. Iron, as an essential trace element, has pleiotropic effects on renal function and the progression of kidney diseases. A two-sample Mendelian randomization (MR) analysis was implemented to determine the potential causal effects between systemic iron status on different kidney diseases. Systemic iron status was represented by four iron-related biomarkers: serum iron, ferritin, transferrin saturation (TfSat), and total iron binding capacity (TIBC). For systemic iron status, 163,511, 246,139, 131,471, and 135,430 individuals were included in the genome-wide association study (GWAS) of serum iron, ferritin, TfSat, and TIBC, respectively. For kidney diseases, 653,143 individuals (15,658 cases and 637,485 controls), 657,076 individuals (8160 cases and 648,916 controls), and 659,320 individuals (10,404 cases and 648,916 controls) were included for immunoglobulin A nephropathy (IgAN), acute kidney disease (AKD), and chronic kidney disease (CKD), respectively. Our MR results showed that increased serum iron [odds ratio (OR): 1.10; 95% confidence interval (95% CI): 1.04, 1.16; p < 0.0042], ferritin (OR: 1.30; 95% CI: 1.14, 1.48; p < 0.0042), and TfSat (OR: 1.07; 95% CI: 1.04, 1.11; p < 0.0042)] and decreased TIBC (OR: 0.92; 95% CI: 0.88, 0.97; p < 0.0042) were associated with elevated IgAN risk. However, no significant associations were found between systemic iron status and AKD or CKD. In our MR study, the genetic evidence supports elevated systemic iron status as a causal effect on IgAN, which suggests a potential protective effect of iron chelation on IgAN patients.
Collapse
Affiliation(s)
- Jiahui Zhou
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wanting Shi
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dongya Wu
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shujie Wang
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xinhui Wang
- Sir Run Run Shaw Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
142
|
Luo F, Huang C. New Insight into Neuropathic Pain: The Relationship between α7nAChR, Ferroptosis, and Neuroinflammation. Int J Mol Sci 2024; 25:6716. [PMID: 38928421 PMCID: PMC11203537 DOI: 10.3390/ijms25126716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/15/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Neuropathic pain, which refers to pain caused by a lesion or disease of the somatosensory system, represents a wide variety of peripheral or central disorders. Treating neuropathic pain is quite demanding, primarily because of its intricate underlying etiological mechanisms. The central nervous system relies on microglia to maintain balance, as they are associated with serving primary immune responses in the brain next to cell communication. Ferroptosis, driven by phospholipid peroxidation and regulated by iron, is a vital mechanism of cell death regulation. Neuroinflammation can be triggered by ferroptosis in microglia, which contributes to the release of inflammatory cytokines. Conversely, neuroinflammation can induce iron accumulation in microglia, resulting in microglial ferroptosis. Accumulating evidence suggests that neuroinflammation, characterized by glial cell activation and the release of inflammatory substances, significantly exacerbates the development of neuropathic pain. By inhibiting microglial ferroptosis, it may be possible to prevent neuroinflammation and subsequently alleviate neuropathic pain. The activation of the homopentameric α7 subtype of the neuronal nicotinic acetylcholine receptor (α7nAChR) has the potential to suppress microglial activation, transitioning M1 microglia to an M2 phenotype, facilitating the release of anti-inflammatory factors, and ultimately reducing neuropathic pain. Recent years have witnessed a growing recognition of the regulatory role of α7nAChR in ferroptosis, which could be a potential target for treating neuropathic pain. This review summarizes the mechanisms related to α7nAChR and the progress of ferroptosis in neuropathic pain according to recent research. Such an exploration will help to elucidate the relationship between α7nAChR, ferroptosis, and neuroinflammation and provide new insights into neuropathic pain management.
Collapse
Affiliation(s)
- Fangting Luo
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China;
| | - Cheng Huang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China;
- Department of Physiology, School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
- Pain Medicine Research Institute, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
143
|
Zhu M, Yuan Z, Wen C, Wei X. DEX Inhibits H/R-induced Cardiomyocyte Ferroptosis by the miR-141-3p/lncRNA TUG1 Axis. Thorac Cardiovasc Surg 2024. [PMID: 38889747 DOI: 10.1055/s-0044-1787691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
BACKGROUND Ferroptosis is emerging as a critical pathway in ischemia/reperfusion (I/R) injury, contributing to compromised cardiac function and predisposing individuals to sepsis and myocardial failure. The study investigates the underlying mechanism of dexmedetomidine (DEX) in hypoxia/reoxygenation (H/R)-induced ferroptosis in cardiomyocytes, aiming to identify novel targets for myocardial I/R injury treatment. METHODS H9C2 cells were subjected to H/R and treated with varying concentrations of DEX. Additionally, H9C2 cells were transfected with miR-141-3p inhibitor followed by H/R treatment. Levels of miR-141-3p, long noncoding RNA (lncRNA) taurine upregulated 1 (TUG1), Fe2+, glutathione (GSH), and malondialdehyde were assessed. Reactive oxygen species (ROS) generation was measured via fluorescent labeling. Expression of ferroptosis-related proteins glutathione peroxidase 4 (GPX4) and acyl-CoA synthetase long-chain family member 4 (ACSL4) was determined using Western blot. The interaction between miR-141-3p and lncRNA TUG1 was evaluated through RNA pull-down assay and dual-luciferase reporter gene assays. The stability of lncRNA TUG1 was assessed using actinomycin D. RESULTS DEX ameliorated H/R-induced cardiomyocyte injury and elevated miR-141-3p expression in cardiomyocytes. DEX treatment increased cell viability, Fe2+, and ROS levels while decreasing ACSL4 protein expression. Furthermore, DEX upregulated GSH and GPX4 protein levels. miR-141-3p targeted lncRNA TUG1, reducing its stability and overall expression. Inhibition of miR-141-3p or overexpression of lncRNA TUG1 partially reversed the inhibitory effect of DEX on H/R-induced ferroptosis in cardiomyocytes. CONCLUSION DEX mitigated H/R-induced ferroptosis in cardiomyocytes by upregulating miR-141-3p expression and downregulating lncRNA TUG1 expression, unveiling a potential therapeutic strategy for myocardial I/R injury.
Collapse
Affiliation(s)
- Mei Zhu
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, People's Republic of China
| | - Zhiguo Yuan
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, People's Republic of China
| | - Chuanyun Wen
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, People's Republic of China
| | - Xiaojia Wei
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, People's Republic of China
| |
Collapse
|
144
|
Fortuna V, Lima J, Oliveira GF, Oliveira YS, Getachew B, Nekhai S, Aschner M, Tizabi Y. Ferroptosis as an emerging target in sickle cell disease. Curr Res Toxicol 2024; 7:100181. [PMID: 39021403 PMCID: PMC11252799 DOI: 10.1016/j.crtox.2024.100181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Sickle cell disease (SCD) is an inherited hemoglobin disorder marked by red blood cell sickling, resulting in severe anemia, painful episodes, extensive organ damage, and shortened life expectancy. In SCD, increased iron levels can trigger ferroptosis, a specific type of cell death characterized by reactive oxygen species (ROS) and lipid peroxide accumulation, leading to damage and organ impairments. The intricate interplay between iron, ferroptosis, inflammation, and oxidative stress in SCD underscores the necessity of thoroughly understanding these processes for the development of innovative therapeutic strategies. This review highlights the importance of balancing the complex interactions among various factors and exploitation of the knowledge in developing novel therapeutics for this devastating disease.
Collapse
Affiliation(s)
- Vitor Fortuna
- Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Jaqueline Lima
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Gabriel F. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Yasmin S. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Sergei Nekhai
- Center for Sickle Cell Disease, Departments of Microbiology and Medicine, Howard University College of Medicine, Washington, DC, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
145
|
Liu Y, Feng D, Shui L, Wang YJ, Yu L, Liu YQ, Tian JY. The research landscape of ferroptosis in neurodegenerative disease: a bibliometric analysis. Front Aging Neurosci 2024; 16:1417989. [PMID: 38962561 PMCID: PMC11221830 DOI: 10.3389/fnagi.2024.1417989] [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: 04/15/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024] Open
Abstract
Background Ferroptosis, a newly proposed concept of programmed cell death, has garnered significant attention in research across different diseases in the last decade. Despite thorough citation analyses in neuroscience, there is a scarcity of information on ferroptosis research specifically related to neurodegenerative diseases. Method The Web of Science Core Collection database retrieved relevant articles and reviews. Data on publications, countries, institutions, authors, journals, citations, and keywords in the included studies were systematically analyzed using Microsoft Excel 2019 and CiteSpace 6.2.R7 software. Result A comprehensive analysis and visualization of 563 research papers on ferroptosis in neurodegenerative diseases from 2014 to 2023 revealed emerging research hotspots and trends. The number of annual publications in this field of study has displayed a pattern of stabilization in the early years of the decade, followed by a notable increase in the later years and peaking in 2023 with 196 publications. Regarding publication volume and total citations, notable research contributions were observed from countries, institutions, and authors in North America, Western Europe, and China. Current research endeavors primarily focus on understanding the intervention mechanisms of neurodegenerative diseases through the ferroptosis pathway and exploring and identifying potential therapeutic targets. Conclusion The study highlights key areas of interest and emerging trends in ferroptosis research on neurodegenerative diseases, offering valuable insights for further exploration and potential directions for diagnosing and treating such conditions.
Collapse
Affiliation(s)
- Yun Liu
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dan Feng
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ling Shui
- Department of General Practice, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yu-jie Wang
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Li Yu
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yu-qi Liu
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jin-yong Tian
- Department of General Practice, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
146
|
Wang C, Zhao H, Liu Y, Qu M, Lv S, He G, Liang H, Chen K, Yang L, He Y, Ou C. Neurotoxicity of manganese via ferroptosis induced by redox imbalance and iron overload. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116404. [PMID: 38705038 DOI: 10.1016/j.ecoenv.2024.116404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/03/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
Manganese (Mn) is an essential trace element for maintaining bodily functions. Excessive exposure to Mn can pose serious health risks to humans and animals, particularly to the nervous system. While Mn has been implicated as a neurotoxin, the exact mechanism of its toxicity remains unclear. Ferroptosis is a form of programmed cell death that results from iron-dependent lipid peroxidation. It plays a role in various physiological and pathological cellular processes and may be closely related to Mn-induced neurotoxicity. However, the mechanism of ferroptosis in Mn-induced neurotoxicity has not been thoroughly investigated. Therefore, this study aims to investigate the role and mechanism of ferroptosis in Mn-induced neurotoxicity. Using bioinformatics, we identified significant changes in genes associated with ferroptosis in Mn-exposed animal and cellular models. We then evaluated the role of ferroptosis in Mn-induced neurotoxicity at both the animal and cellular levels. Our findings suggest that Mn exposure causes weight loss and nervous system damage in mice. In vitro and in vivo experiments have shown that exposure to Mn increases malondialdehyde, reactive oxygen species, and ferrous iron, while decreasing glutathione and adenosine triphosphate. These findings suggest that Mn exposure leads to a significant increase in lipid peroxidation and disrupts iron metabolism, resulting in oxidative stress injury and ferroptosis. Furthermore, we assessed the expression levels of proteins and mRNAs related to ferroptosis, confirming its significant involvement in Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Changyong Wang
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Hongyan Zhao
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Yaoyang Liu
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Minghai Qu
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Shanyu Lv
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Guoguo He
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Hongshuo Liang
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Kemiao Chen
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Lin Yang
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Yonghua He
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Chaoyan Ou
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, Guilin Medical University, Guilin, Guangxi 541199, China; School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| |
Collapse
|
147
|
Chi LK, Yuan Q, Wang MY, Guo CR, Zhu XD, Jiang HB, Zhang QH, Zhao Y, Li L, Yan H. Metabolomics reveals that ferroptosis participates in bisphenol A-induced testicular injury. Heliyon 2024; 10:e31667. [PMID: 38882385 PMCID: PMC11177062 DOI: 10.1016/j.heliyon.2024.e31667] [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: 10/05/2023] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Objective Bisphenol A (BPA) is a common environmental endocrine disruptor that negatively impairs male reproductive ability. This study aimed to explore the alterations in serum metabolomics that occur following BPA exposure and the mechanism via which BPA induces the death of testicular cells in a male mouse model. Methods The mice were classified into two groups: BPA-exposed and control groups, and samples were collected for metabolomic determination, semen quality analysis, electron microscopy, enzyme-linked immunosorbent assay, quantitative real-time PCR, pathological staining, and Western blot analysis. Results BPA exposure caused testicular damage and significantly decreased sperm quality in mice. Combined with non-target metabolomic analysis, this was closely related to ferroptosis induced by abnormal metabolites of arachidonic acid and phosphatidylcholine, and the expression of its related genes, acyl CoA synthetase 4, glutathione peroxidase 4, lysophosphatidylcholine acyltransferase 3, and phosphatidylethanolamine-binding protein 1 were altered. Conclusion BPA induced ferroptosis, caused testicular damage, and reduced fertility by affecting lipid metabolism in male mice. Inhibiting ferroptosis may potentially function as a therapeutic strategy to mitigate the male reproductive toxicity induced by BPA.
Collapse
Affiliation(s)
- Ling Kan Chi
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qing Yuan
- Department of Gynecology, Department of Fetal Medicine and Prenatal Diagnosis Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201203, China
| | - Min Yan Wang
- Department of Pathology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chun Rong Guo
- Teaching Experimental Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xian Dan Zhu
- Laboratory Animal Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hua Bo Jiang
- Department of Gynecology, Department of Fetal Medicine and Prenatal Diagnosis Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201203, China
| | - Qin Hua Zhang
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuan Zhao
- Laboratory Animal Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Li
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hua Yan
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
148
|
Yu X, Wang S, Ji Z, Meng J, Mou Y, Wu X, Yang X, Xiong P, Li M, Guo Y. Ferroptosis: An important mechanism of disease mediated by the gut-liver-brain axis. Life Sci 2024; 347:122650. [PMID: 38631669 DOI: 10.1016/j.lfs.2024.122650] [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: 02/22/2024] [Revised: 03/27/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024]
Abstract
AIMS As a unique iron-dependent non-apoptotic cell death, Ferroptosis is involved in the pathogenesis and development of many human diseases and has become a research hotspot in recent years. However, the regulatory role of ferroptosis in the gut-liver-brain axis has not been elucidated. This paper summarizes the regulatory role of ferroptosis and provides theoretical basis for related research. MATERIALS AND METHODS We searched PubMed, CNKI and Wed of Science databases on ferroptosis mediated gut-liver-brain axis diseases, summarized the regulatory role of ferroptosis on organ axis, and explained the adverse effects of related regulatory effects on various diseases. KEY FINDINGS According to our summary, the main way in which ferroptosis mediates the gut-liver-brain axis is oxidative stress, and the key cross-talk of ferroptosis affecting signaling pathway network is Nrf2/HO-1. However, there were no specific marker between different organ axes mediate by ferroptosis. SIGNIFICANCE Our study illustrates the main ways and key cross-talk of ferroptosis mediating the gut-liver-brain axis, providing a basis for future research.
Collapse
Affiliation(s)
- Xinxin Yu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Shihao Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Zhongjie Ji
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Jiaqi Meng
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Yunying Mou
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Xinyi Wu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Xu Yang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Panyang Xiong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Mingxia Li
- Nursing School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Yinghui Guo
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China.
| |
Collapse
|
149
|
Du Z, Shi Y, Tan J. Advances in integrating single-cell sequencing data to unravel the mechanism of ferroptosis in cancer. Brief Funct Genomics 2024:elae025. [PMID: 38874174 DOI: 10.1093/bfgp/elae025] [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: 03/18/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
Ferroptosis, a commonly observed type of programmed cell death caused by abnormal metabolic and biochemical mechanisms, is frequently triggered by cellular stress. The occurrence of ferroptosis is predominantly linked to pathophysiological conditions due to the substantial impact of various metabolic pathways, including fatty acid metabolism and iron regulation, on cellular reactions to lipid peroxidation and ferroptosis. This mode of cell death serves as a fundamental factor in the development of numerous diseases, thereby presenting a range of therapeutic targets. Single-cell sequencing technology provides insights into the cellular and molecular characteristics of individual cells, as opposed to bulk sequencing, which provides data in a more generalized manner. Single-cell sequencing has found extensive application in the field of cancer research. This paper reviews the progress made in ferroptosis-associated cancer research using single-cell sequencing, including ferroptosis-associated pathways, immune checkpoints, biomarkers, and the identification of cell clusters associated with ferroptosis in tumors. In general, the utilization of single-cell sequencing technology has the potential to contribute significantly to the investigation of the mechanistic regulatory pathways linked to ferroptosis. Moreover, it can shed light on the intricate connection between ferroptosis and cancer. This technology holds great promise in advancing tumor-wide diagnosis, targeted therapy, and prognosis prediction.
Collapse
Affiliation(s)
- Zhaolan Du
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Yi Shi
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Jianjun Tan
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
150
|
Luo Y, Zhang Z, Xi S, Li T. Bioinformatics analyses and experimental validation of ferroptosis-related genes in bronchopulmonary dysplasia pathogenesis. PLoS One 2024; 19:e0291583. [PMID: 38875180 PMCID: PMC11178182 DOI: 10.1371/journal.pone.0291583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 05/18/2024] [Indexed: 06/16/2024] Open
Abstract
OBJECTIVE We aimed to study the involvement of ferroptosis in the pathogenesis of bronchopulmonary dysplasia (BPD) by conducting bioinformatics analyses and identifying and validating the associated ferroptosis-related genes to explore new directions for treating BPD. METHODS The dataset GSE32472 on BPD was downloaded from the public genome database. Using R language, differentially expressed genes (DEGs) between the BPD and normal group were screened. In the present study, we adopted weighted gene correlation network analysis (WGCNA) for identifying BPD-related gene modules and ferroptosis-related genes were extracted from FerrDb. Their results were intersected to obtain the hub genes. After that, to explore the hub gene-related signaling pathways, the hub genes were exposed to gene ontology enrichment analysis. With the purpose of verifying the mRNA expression of the hub genes, a single-gene gene set enrichment analysis and quantitative reverse transcription polymerase chain reaction were conducted. Immune cell infiltration in BPD was analyzed using the CIBERSORT inverse fold product algorithm. RESULTS A total of 606 DEGs were screened. WGCNA provided the BPD-related gene module darkgreen4. The intersection of DEGs, intramodular genes, and ferroptosis-related genes revealed six ferroptosis-associated hub genes (ACSL1, GALNT14, WIPI1, MAPK14, PROK2, and CREB5). Receiver operating characteristic curve analysis demonstrated that the hub genes screened for BPD were of good diagnostic significance. According to the results of immune infiltration analysis, the proportions of CD8, CD4 naive, and memory resting T cells and M2 macrophage were elevated in the normal group, and the proportions of M0 macrophage, resting mast cell, and neutrophils were increased in the BPD group. CONCLUSIONS A total of six ferroptosis-associated hub genes in BPD were identified in this study, and they may be potential new therapeutic targets for BPD.
Collapse
Affiliation(s)
- Yifan Luo
- Department of Pediatrics, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Zongli Zhang
- Institute of Pediatric Diseases, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Shibing Xi
- Department of Pediatrics, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Tao Li
- Department of Pediatrics, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|