1
|
Wang L, Yu H, Wang D, Yin G, Chen S, Zhang X, Yu W, Meng D, Liu H, Jiang W, Zhang F. Diosgenin alleviates lipid accumulation in NAFLD through the pathways of ferroptosis defensive and executive system. J Nutr Biochem 2025; 140:109886. [PMID: 40023201 DOI: 10.1016/j.jnutbio.2025.109886] [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: 08/26/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
The most prevalent liver condition globally is non-alcoholic fatty liver disease (NAFLD), for which no approved therapies currently exist. Diosgenin, an important component in plants from the Leguminosae, Dioscoreaceae, and Solanaceae families, has demonstrated considerable anti-inflammatory and antioxidant effects. Nonetheless, the specific mechanism by which it may act in managing NAFLD remains unclear. Our research aims to explore the effects and molecular mechanisms of DG on NAFLD by utilizing both in vivo and in vitro experimental approaches. To investigate the effect of DG on hepatic steatosis, we used Sprague-Dawley rats induced by a high-fat diet (HFD) and HepG2 cells exposed to free fatty acids. Oil red O staining and hematoxylin-eosin (H&E) staining were used to explore lipid accumulation and hepatic degeneration. ROS staining, SOD, MDA, and Fe2+kits were used to detect the indexes related to oxidative stress in ferroptosis in hepatic tissues and cells. IFSP1 and pcDNA3.1-ACSL4 plasmid were used to knock down Ferroptosis suppressor protein1 (FSP1) and promote the expression of acyl-CoA synthetase long-chain family member 4 (ACSL4) in HepG2 cells. DG improved lipid metabolism disorders and liver damage induced by a high-fat diet in rats with NAFLD. Furthermore, the administration of DG notably decreased oxidative stress levels and liver Fe2+ concentrations in rats. Additionally, in vitro experiments demonstrated that DG treatment markedly attenuated ferroptosis and ROS accumulation in HepG2 cells induced by FFAs. Moreover, overexpression of hepatic ACSL4 expression by pcDNA3.1-ACSL4 plasmid promoted the regulatory effects of DG on LPCAT3 and ALOX15. Our research shows that DG can alleviate NAFLD by regulating the FSP1/COQ10 pathway of the ferroptosis defense system and the ACSL4/LPCAT3/ALOX15 pathway of the ferroptosis execution system. Therefore, DG may serve as a novel inhibitor of ferroptosis for the treatment of NAFLD.
Collapse
Affiliation(s)
- Linya Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongzhuan Yu
- Weifang Traditional Chinese Medicine Hospital, Shandong, China
| | - Dongxian Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Suwen Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfei Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Decheng Meng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongshuai Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenying Jiang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fengxia Zhang
- Department of Neurology, the first Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, China.
| |
Collapse
|
2
|
Yu Y, Yang Y, Li Q, Yuan J, Zha Y. Predicting metabolic dysfunction associated steatotic liver disease using explainable machine learning methods. Sci Rep 2025; 15:12382. [PMID: 40216893 PMCID: PMC11992218 DOI: 10.1038/s41598-025-96478-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Early and accurate identification of patients at high risk of metabolic dysfunction-associated steatotic liver disease (MASLD) is critical to prevent and improve prognosis potentially. We aimed to develop and validate an explainable prediction model based on machine learning (ML) approaches for MASLD among the adult population. The national cross-sectional study collected data from the National Health and Nutrition Examination Survey from 2017 to 2020, consisting of 13,436 participants, who were randomly split into 70% training, 20% internal validation, and 10% external validation cohorts. MASLD was defined based on transient elastography and cardiometabolic risk factors. With 50 medical characteristics easily obtained, six ML algorithms were used to develop prediction models. Several evaluation parameters were used to compare the predictive performance, including the area under the receiver-operating-characteristic curve (AUC) and precision-recall (P-R) curve. The recursive feature elimination method was applied to select the optimal feature subset. The Shapley Additive exPlanations method offered global and local explanations for the model. The random forest (RF) model performed best in discriminative ability among 6 ML models, and the optimal 10-feature RF model was finally chosen. The final model could accurately predict MASLD in internal and external validation cohorts (AUC: 0.928, 0.918; area under P-R curve: 0.876, 0.863, respectively). The final model performed better than each of the traditional risk indicators for MASLD. An explainable 10-feature prediction model with excellent discrimination and calibration performance was successfully developed and validated for MASLD based on clinical data easily extracted using an RF algorithm.
Collapse
Affiliation(s)
- Yihao Yu
- Master of Finance, Australian National University, Canberra, Australia
| | - Yuqi Yang
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, 550002, China
- NHC Key Laboratory of Pulmonary Immunological Disease, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Qian Li
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, 550002, China
- NHC Key Laboratory of Pulmonary Immunological Disease, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Jing Yuan
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, 550002, China
- NHC Key Laboratory of Pulmonary Immunological Disease, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Yan Zha
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
- NHC Key Laboratory of Pulmonary Immunological Disease, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| |
Collapse
|
3
|
Li L, Guo Z, Zhao Y, Liang C, Zheng W, Tian W, Chen Y, Cheng Y, Zhu F, Xiang X. The impact of oxidative stress on abnormal lipid metabolism-mediated disease development. Arch Biochem Biophys 2025; 766:110348. [PMID: 39961502 DOI: 10.1016/j.abb.2025.110348] [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: 11/06/2024] [Revised: 02/09/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Oxidative stress arises from an imbalance between cellular oxidation and anti-oxidation mechanisms, leading to various harmful effects on physiological health. These include inflammatory neutrophil infiltration, increased secretion of proteases, and increased production of oxidative intermediates, all of which significantly contribute to aging and the onset of multiple diseases. This review explores abnormal lipid metabolism, characterized by dysregulation in lipid synthesis, catabolism, digestion, absorption, and transport, with the potential to lead to lipid droplet accumulation or deficit across tissues, thus causing adverse health outcomes. Importantly, the intricate relationship between oxidative stress and inflammation plays a central role in exacerbating metabolic disorders, including diabetes, obesity, hypertension, non-alcoholic fatty liver disease, atherosclerosis, and lung fibrosis. This review seeks to compile and integrate recent research findings on the influence of oxidative stress on abnormal lipid metabolism pathology. A deeper understanding of this connection could reveal new perspectives for advancing the treatment and management of metabolic disorders.
Collapse
Affiliation(s)
- Lanlan Li
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Zhiliang Guo
- The 80th Group Army Hospital of Chinese PLA, Weifang, Shandong, 261021, China
| | - Yi Zhao
- Shandong Provincial Hospital Affiliated with Shandong's First Medical University, Shandong, China
| | - Chuanjie Liang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Wenxiang Zheng
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Wenxiu Tian
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Yalin Chen
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Yi Cheng
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Fengwen Zhu
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255000, Shandong, China.
| | - Xinxin Xiang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255000, Shandong, China.
| |
Collapse
|
4
|
Chen KQ, Wang SZ, Lei HB, Liu X. The Relationship Between Ferroptosis and Metabolic Dysfunction-Associated Fatty Liver Disease. Liver Int 2025; 45:e16135. [PMID: 39422264 DOI: 10.1111/liv.16135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Ke-Qian Chen
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, China
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, China
| | - Hai-Bo Lei
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, China
| |
Collapse
|
5
|
Wang S, Du R, Liu J, Zhong W, Zhang C, Jiang X, Wang X, Wu Q, Tong G, Luo L. Multi-approach analysis reveals the mechanism by which Shugan Xiaozhi decoction protects against metabolic dysfunction-associated steatohepatitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156712. [PMID: 40220418 DOI: 10.1016/j.phymed.2025.156712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/08/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) is a human health-threatening hepatic disease with limited treatment strategies. As a clinical Traditional Chinese Medicine compound for MASH, Shugan Xiaozhi (SGXZ) decoction has a definite effect, but its mechanism in treating MASH is still not very clear. PURPOSE Exploring the potential mechanism of SGXZ decoction in treating MASH through multiomics and animal experimental validation. METHODS UPLC-ESI-MS method was used to identify the main components of SGXZ decoction. Periodic acid-schiff (PAS), picrosirius red (PSR), and oil red o staining were used to assess the effect of SGXZ decoction on MCD-induced MASH mouse model. The mechanism of SGXZ decoction on MASH was analyzed using multiomics techniques. TUNEL staining, western blot (WB), immunohistochemistry (IHC), kits, transmission electron microscopy (TEM), and immunofluorescence (IF) were used to validate the mechanism of SGXZ decoction on MASH. Finally, molecular docking and molecular dynamics simulation were used to verify the targeting between key components of SGXZ decoction and important targets for intervention. RESULTS Through UPLC-ESI-MS analysis, 30 main active ingredients were obtained from SGXZ decoction. SGXZ decoction improved MASH, as evidenced by the improvement in histopathology, hepatic function indexes, lipid and fibrosis indicators. Both proteomic and transcriptomic results suggested an important role for ferroptosis in SGXZ decoction intervention in MASH, ferroptosis-related pathways were the main significant pathways obtained from these analyses. In addition, SGXZ decoction treatment reduced cell death, inflammation, and oxidative stress levels and restored impaired mitochondrial morphology in MCD-induced MASH mice. Furthermore, Mechanism experiments proved that SGXZ decoction treatment improved iron metabolism and lipid peroxidation imbalance and activated the Xc- system in MASH mice. CONCLUSION SGXZ decoction does have a therapeutic effect on MASH, and its mechanism may be related to its regulation of p53/ SLC7A11/GPX4 pathway to reduce ferroptosis.
Collapse
Affiliation(s)
- Shuai Wang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Ruili Du
- The First Clinical Medical College of Henan University of Chinese Medicine, No. 19, Renmin Road, Jinshui District, Henan, 450003, PR China
| | - Jiahui Liu
- Department of Nephrology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong 518033, PR China
| | - Weichao Zhong
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Chunmei Zhang
- School of Basic Medical Science of Luoyang Polytechnic, No. 6 Keji Avenue, Yibin District, Henan, 471099, PR China
| | - Xia Jiang
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, PR China
| | - Xiaohui Wang
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao ln-Depth Cooperation Zone in Hengqin, 519000, PR China.
| | - Guangdong Tong
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China.
| | - Lidan Luo
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China.
| |
Collapse
|
6
|
Matboli M, Hamady S, Saad M, Khaled R, Khaled A, Barakat EMF, Sayed SA, Agwa S, Youssef I. Innovative approaches to metabolic dysfunction-associated steatohepatitis diagnosis and stratification. Noncoding RNA Res 2025; 10:206-222. [PMID: 40248839 PMCID: PMC12004009 DOI: 10.1016/j.ncrna.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/08/2024] [Accepted: 10/10/2024] [Indexed: 01/03/2025] Open
Abstract
The global rise in Metabolic dysfunction-associated steatotic liver disease (MASLD)/Metabolic dysfunction-associated steatohepatitis (MASH) highlights the urgent necessity for noninvasive biomarkers to detect these conditions early. To address this, we endeavored to construct a diagnostic model for MASLD/MASH using a combination of bioinformatics, molecular/biochemical data, and machine learning techniques. Initially, bioinformatics analysis was employed to identify RNA molecules associated with MASLD/MASH pathogenesis and enriched in ferroptosis and exophagy. This analysis unveiled specific networks related to ferroptosis (GPX4, LPCAT3, ACSL4, miR-4266, and LINC00442) and exophagy (TSG101, HGS, SNF8, miR-4498, miR-5189-5p, and CTBP1-AS2). Subsequently, serum samples from 400 participants (151 healthy, 150 MASH, and 99 MASLD) underwent biochemical and molecular analysis, revealing significant dyslipidemia, impaired liver function, and disrupted glycemic indicators in MASLD/MASH patients compared to healthy controls. Molecular analysis indicated increased expression of LPCAT3, ACSL4, TSG101, HGS, and SNF8, alongside decreased GPX4 levels in MASH and MASLD patients compared to controls. The expression of epigenetic regulators from both networks (miR-4498, miR-5189-5p, miR-4266, LINC00442, and CTBP1-AS2) significantly differed among the studied groups. Finally, supervised machine learning models, including Neural Networks and Random Forest, were applied to molecular signatures and clinical/biochemical data. The Random Forest model exhibited superior performance, and molecular features effectively distinguished between the three studied groups. Clinical features, particularly BMI, consistently served as discriminatory factors, while biochemical features exhibited varying discriminant behavior across MASH, MASLD, and control groups. Our study underscores the significant potential of integrating diverse data types to enable early detection of MASLD/MASH, offering a promising approach for non-invasive diagnostic strategies.
Collapse
Affiliation(s)
- Marwa Matboli
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt
- Faculty of Oral & Dental Medicine, Misr International University, Qalyubiyya Governorate, Egypt
| | - Shaimaa Hamady
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Maha Saad
- Basic Sciences Department, Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| | - Radwa Khaled
- Basic Sciences Department, Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University & Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| | - Abdelrahman Khaled
- Bioinformatics Group, Center of Informatics Sciences (CIS), School of Information Technology and Computer Sciences, Nile University, Giza, Egypt
| | - Eman MF. Barakat
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sayed Ahmed Sayed
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - SaraH.A. Agwa
- Clinical Pathology and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo, 11382, Egypt
| | - Ibrahim Youssef
- Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Egypt
| |
Collapse
|
7
|
Ma MJ, Lin J, Yang HJ, You J. Relationship between iron and lipid peroxidation in ferroptosis and effect of ferroptosis in metabolic dysfunction-associated steatotic liver disease. Shijie Huaren Xiaohua Zazhi 2025; 33:28-36. [DOI: 10.11569/wcjd.v33.i1.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 01/25/2025] Open
Abstract
The liver plays an irreplaceable role in human body functions, and liver damage of various causes is a major problem that plagues human health. China is a country with a heavy burden of hepatitis B, but in recent years, the prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has shown an increasing trend. Although the mechanism of liver injury caused by MASLD is not completely clear, it is inextricably related to the body's metabolism. MASLD is one of the most common chronic liver diseases and is considered to be a manifestation of metabolic syndrome in the liver. Ferroptosis is a cell death mechanism discovered in recent years, which is characterized by iron metabolism disorders and lipid peroxide accumulation. In recent years, several studies have found that there is an inextricable relationship between ferroptosis and liver disease. This article describes the relationship between iron or iron homeostasis and lipid peroxidation from the perspective of iron metabolism disorders, and the effect of ferroptosis in MASLD.
Collapse
Affiliation(s)
- Meng-Juan Ma
- Institute of Geriatric Medicine, Clinical Research Center for Geriatric Diseases, Department of Geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Jie Lin
- Institute of Geriatric Medicine, Clinical Research Center for Geriatric Diseases, Department of Geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Hong-Ju Yang
- Institute of Geriatric Medicine, Clinical Research Center for Geriatric Diseases, Department of Geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Jing You
- Institute of Geriatric Medicine, Clinical Research Center for Geriatric Diseases, Department of Geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| |
Collapse
|
8
|
Ma MJ, Lin J, Yang HJ, You J. Relationship between iron and lipid peroxidation in ferroptosis and effect of ferroptosis in metabolic dysfunction-associated steatotic liver disease. Shijie Huaren Xiaohua Zazhi 2025; 33:34-42. [DOI: 10.11569/wcjd.v33.i1.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
The liver plays an irreplaceable role in human body functions, and liver damage of various causes is a major problem that plagues human health. China is a country with a heavy burden of hepatitis B, but in recent years, the prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has shown an increasing trend. Although the mechanism of liver injury caused by MASLD is not completely clear, it is inextricably related to the body's metabolism. MASLD is one of the most common chronic liver diseases and is considered to be a manifestation of metabolic syndrome in the liver. Ferroptosis is a cell death mechanism discovered in recent years, which is characterized by iron metabolism disorders and lipid peroxide accumulation. In recent years, several studies have found that there is an inextricable relationship between ferroptosis and liver disease. This article describes the relationship between iron or iron homeostasis and lipid peroxidation from the perspective of iron metabolism disorders, and the effect of ferroptosis in MASLD.
Collapse
Affiliation(s)
- Meng-Juan Ma
- Institute of Geriatric Medicine, Clinical Research Center for Geriatric Diseases, Department of Geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Jie Lin
- Institute of Geriatric Medicine, Clinical Research Center for Geriatric Diseases, Department of Geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Hong-Ju Yang
- Institute of Geriatric Medicine, Clinical Research Center for Geriatric Diseases, Department of Geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Jing You
- Institute of Geriatric Medicine, Clinical Research Center for Geriatric Diseases, Department of Geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| |
Collapse
|
9
|
Ge T, Wang Y, Han Y, Bao X, Lu C. Exploring the Updated Roles of Ferroptosis in Liver Diseases: Mechanisms, Regulators, and Therapeutic Implications. Cell Biochem Biophys 2024:10.1007/s12013-024-01611-3. [PMID: 39543068 DOI: 10.1007/s12013-024-01611-3] [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: 10/29/2024] [Indexed: 11/17/2024]
Abstract
Ferroptosis, a newly discovered mode of cell death, is a type of iron-dependent regulated cell death characterized by intracellular excessive lipid peroxidation and imbalanced redox. As the liver is susceptible to oxidative damage and the abnormal iron accumulation is a major feature of most liver diseases, studies on ferroptosis in the field of liver diseases are of great interest. Studies show that targeting the key regulators of ferroptosis can effectively alleviate or even reverse the deterioration process of liver diseases. System Xc- and glutathione peroxidase 4 are the main defense regulators of ferroptosis, while acyl-CoA synthetase long chain family member 4 is a key enzyme causing peroxidation in ferroptosis. Generally speaking, ferroptosis should be suppressed in alcoholic liver disease, non-alcoholic fatty liver disease, and drug-induced liver injury, while it should be induced in liver fibrosis and hepatocellular carcinoma. In this review, we summarize the main regulators involved in ferroptosis and then the mechanisms of ferroptosis in different liver diseases. Treatment options of drugs targeting ferroptosis are further concluded. Determining different triggers of ferroptosis can clarify the mechanism of ferroptosis occurs at both physiological and pathological levels.
Collapse
Affiliation(s)
- Ting Ge
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yang Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yiwen Han
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
10
|
Maddie N, Chacko N, Matatov D, Carrillo‐Sepulveda MA. Western diet promotes the progression of metabolic dysfunction-associated steatotic liver disease in association with ferroptosis in male mice. Physiol Rep 2024; 12:e70139. [PMID: 39610053 PMCID: PMC11604596 DOI: 10.14814/phy2.70139] [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: 06/10/2024] [Revised: 10/29/2024] [Accepted: 11/16/2024] [Indexed: 11/30/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), now referred to as metabolic dysfunction-associated steatotic liver disease (MASLD), is a silent killer that often progresses to metabolic dysfunction-associated steatohepatitis (MASH). To date, there are no pharmacological treatments for MASLD. While obesity is a major cause of the development and progression of MASLD, the underlying mechanisms remain unclear. We hypothesize that ferroptosis, a recently discovered nonapoptotic iron-dependent form of cell death, is activated during the progression of MASLD and may be a potential target for treating MASLD. Using a murine model of Western diet-induced obesity, C57BL/6J male mice were exposed to a long-term (36 weeks) Western diet. Controls were maintained with a standard chow diet. Western diet-induced obesity was confirmed by increased body mass index (BMI). Histopathological analysis demonstrated the progression of MASLD to MASH in the obese group, which was accompanied by significant hepatic iron deposition, oxidative damage, and lipid peroxidation. Hepatic ferroptosis was further confirmed by decreased protein expression of glutathione peroxidase 4 (GPX4) and increased acyl-CoA synthetase long-chain family member 4 (ACSL4), markers of ferroptosis. These findings suggest that ferroptosis is a potential mechanism involved in the pathogenesis of MASLD in male mice.
Collapse
Affiliation(s)
- Nicole Maddie
- Department of Biomedical Sciences, College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Nefia Chacko
- Department of Biomedical Sciences, College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - David Matatov
- Department of Biomedical Sciences, College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
| | | |
Collapse
|
11
|
Park GC, Bang SY, Kim JM, Shin SC, Cheon YI, Kim KM, Park H, Sung ES, Lee M, Lee JC, Lee BJ. Inhibiting Ferroptosis Prevents the Progression of Steatotic Liver Disease in Obese Mice. Antioxidants (Basel) 2024; 13:1336. [PMID: 39594477 PMCID: PMC11590881 DOI: 10.3390/antiox13111336] [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: 10/17/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Ferroptosis, a form of regulated cell death characterized by lipid peroxidation and iron accumulation, has been implicated in the progression of metabolic-dysfunction-associated steatohepatitis (MASH) in obesity. This study investigated the role of ferroptosis in the development of hepatic steatosis and MASH in obese mice and assessed the therapeutic potential of ferrostatin-1, a ferroptosis inhibitor. C57BL/6J wild-type (n = 8) and ob/ob mice (n = 16) were maintained on a standard chow diet. Mice were divided into three groups that included C57BL/6 (n = 8), ob/ob (n = 8), and ob/ob + ferrostatin-1 (FER) (n = 8), with the latter group receiving an intraperitoneal injection of 5 μM/kg ferrostatin three times per week for eight weeks. Following treatment, serum and tissue samples were collected for analysis. Significant hepatic steatosis and increased lipogenesis markers were observed in ob/ob mice, which were restored to baseline levels in the ob/ob + FER group treated with ferrostatin-1. Elevated oxidative stress was indicated by increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels in the ob/ob group, while glutathione peroxidase 4 (GPX4) activity was significantly reduced. Ferrostatin-1 treatment decreases MDA levels and restores GPX4 activity. Additionally, ferrostatin mitigates iron overload and promotes macrophage polarization from M1 to M2, thereby reducing liver inflammation and fibrosis. Ferrostatin treatment reversed mitochondrial dysfunction in ob/ob mice. Our findings revealed that ferroptosis plays a significant role in the progression of obesity to hepatic steatosis and MASH. Inhibiting ferroptosis using ferrostatin-1 effectively improves liver histology, reduces oxidative stress, normalizes lipogenesis, and modulates macrophage polarization. This study highlights the potential of targeting ferroptosis as a therapeutic strategy for obesity-related liver diseases, warranting further investigation in clinical settings.
Collapse
Affiliation(s)
- Gi Cheol Park
- Department of Otolaryngology—Head and Neck Surgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea; (G.C.P.); (H.P.)
| | - Soo-Young Bang
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.-Y.B.); (J.M.K.); (S.-C.S.); (Y.-i.C.)
| | - Ji Min Kim
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.-Y.B.); (J.M.K.); (S.-C.S.); (Y.-i.C.)
| | - Sung-Chan Shin
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.-Y.B.); (J.M.K.); (S.-C.S.); (Y.-i.C.)
| | - Yong-il Cheon
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.-Y.B.); (J.M.K.); (S.-C.S.); (Y.-i.C.)
| | - Kwang Min Kim
- Division of Gastroenterology, Department of Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea;
| | - Hanaro Park
- Department of Otolaryngology—Head and Neck Surgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea; (G.C.P.); (H.P.)
| | - Eui-Suk Sung
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (E.-S.S.); (M.L.); (J.-C.L.)
| | - Minhyung Lee
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (E.-S.S.); (M.L.); (J.-C.L.)
| | - Jin-Choon Lee
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (E.-S.S.); (M.L.); (J.-C.L.)
| | - Byung-Joo Lee
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.-Y.B.); (J.M.K.); (S.-C.S.); (Y.-i.C.)
| |
Collapse
|
12
|
Xiao Z, Zhou J, Chen H, Chen X, Wang L, Liu D, Kang X. Synthesis, characterization and MAFLD prevention potential of Ganoderma lucidum spore polysaccharide-stabilized selenium nanoparticles. Int J Biol Macromol 2024; 282:136962. [PMID: 39490485 DOI: 10.1016/j.ijbiomac.2024.136962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
The unstability of selenium nanoparticles (SeNPs) results in decreased activity which limits its therapeutic potential. In this study, we utilized Ganoderma lucidum spore polysaccharide (GLP, Mw = 983.96 kDa) as a novel stabilizer to synthesize GLP-SeNPs. GLP-SeNPs (Se/GLP = 1/3) with an average diameter of 149 nm were successfully prepared and it was stable for at least 30 days at 4 °C. It exhibited an orange-red color, zero valence state, amorphous structure, selenium uniform distribution, a zeta potential of -29.73 mV, selenium content of 16.04 %. GLP-SeNPs pretreatment decreased lipid accumulation, reduced ROS content and enhanced SOD and CAT activity in HepG2 cells. Fe2+ and MDA contents were decreased, while GPX4 and GSH activities were increased. All these ameliorated effects could be abolished by NRF2 antagonist ML385. The expression of anti-oxidant genes and iron exporter was up-regulated, while that of pro-oxidant and lipid biosynthesis gene was down-regulated. The GPX4 activity could be reduced by ML385 addition. In conclusion, GLP-SeNPs was successfully constructed at the ratio of 1/3 (Se/GLP). It prevents MAFLD by targeting ferroptosis, including lowering iron overload, inhibiting lipid accumulation and attenuating oxidative stress. The improvement was conducted via activating SLC40A1-mediated iron pathway, ACSL4-mediated lipid metabolism and NRF2-mediated GSH-GPX4 pathway. Therefore, GLP-SeNPs can be used as potential selenium nutritional supplements or adjuvants for MAFLD prevention.
Collapse
Affiliation(s)
- Zhengpeng Xiao
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China
| | - Jiali Zhou
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China
| | - Hanqi Chen
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China
| | - Xuan Chen
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China; Hunan Provincial Engineering Research Center of Medical Nutrition Intervention Technology for Metabolic Diseases, Hunan Agricultural University, Changsha, Hunan, PR China; Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, PR China
| | - Lei Wang
- State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China; Changsha Nengfeng Biotechnology Co., Ltd, Changsha, Hunan, PR China
| | - Dongbo Liu
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China; Hunan Provincial Engineering Research Center of Medical Nutrition Intervention Technology for Metabolic Diseases, Hunan Agricultural University, Changsha, Hunan, PR China; Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, PR China.
| | - Xincong Kang
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China; Hunan Provincial Engineering Research Center of Medical Nutrition Intervention Technology for Metabolic Diseases, Hunan Agricultural University, Changsha, Hunan, PR China; Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, PR China.
| |
Collapse
|
13
|
Peng J, Dai X, Zhang T, Hu G, Cao H, Guo X, Fan H, Chen J, Tang W, Yang F. Copper as the driver of the lncRNA-TCONS-6251/miR-novel-100/TC2N axis: Unraveling ferroptosis in duck kidney. Int J Biol Macromol 2024; 282:136797. [PMID: 39454920 DOI: 10.1016/j.ijbiomac.2024.136797] [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: 07/17/2024] [Revised: 10/08/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
Ferroptosis is an iron-dependent form of oxidative cell death. Competitive endogenous RNAs diminish the inhibitory impact of microRNAs on other transcripts by chelating effects, which affects ferroptosis and reactive oxygen species (ROS) levels. However, the role of ferroptosis in excessive copper (Cu)-induced renal injury via the ceRNA axis has not been fully illustrated yet. Herein, we found that Cu induced ferroptosis in duck renal tubular epithelial cells, as indicated by the increase in intracellular iron levels and lipid peroxidation, upregulation of PTGS2 and ACSL4 levels, reduced GPX4 and GSH levels. In addition, knockdown miR-novel-100 could effectively decreased ferroptosis induced by Cu. Overexpression of miR-novel-100 or TC2N knockdown resulted in the stimulation of ROS and the upregulation of ferroptosis indicators. However, butylated hydroxyanisole (BHA) decreased the stimulation of ROS and the ferroptosis effect caused by miR-novel-100 overexpression. In conclusion, Cu induced ferroptosis by activating the lncRNA-TCONS-6251/miR-novel-100/TC2N axis to cause ROS accumulation.
Collapse
Affiliation(s)
- Junjun Peng
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Xueyan Dai
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Tao Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Huiqin Fan
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Jing Chen
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Wei Tang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Fan Yang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China.
| |
Collapse
|
14
|
Cheng Z, Chu H, Seki E, Lin R, Yang L. Hepatocyte programmed cell death: the trigger for inflammation and fibrosis in metabolic dysfunction-associated steatohepatitis. Front Cell Dev Biol 2024; 12:1431921. [PMID: 39071804 PMCID: PMC11272544 DOI: 10.3389/fcell.2024.1431921] [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: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
By replacing and removing defective or infected cells, programmed cell death (PCD) contributes to homeostasis maintenance and body development, which is ubiquitously present in mammals and can occur at any time. Besides apoptosis, more novel modalities of PCD have been described recently, such as necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death. PCD not only regulates multiple physiological processes, but also participates in the pathogenesis of diverse disorders, including metabolic dysfunction-associated steatotic liver disease (MASLD). MASLD is mainly classified into metabolic dysfunction-associated steatotic liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), and the latter putatively progresses to cirrhosis and hepatocellular carcinoma. Owing to increased incidence and obscure etiology of MASH, its management still remains a tremendous challenge. Recently, hepatocyte PCD has been attracted much attention as a potent driver of the pathological progression from MASL to MASH, and some pharmacological agents have been proved to exert their salutary effects on MASH partly via the regulation of the activity of hepatocyte PCD. The current review recapitulates the pathogenesis of different modalities of PCD, clarifies the mechanisms underlying how metabolic disorders in MASLD induce hepatocyte PCD and how hepatocyte PCD contributes to inflammatory and fibrotic progression of MASH, discusses several signaling pathways in hepatocytes governing the execution of PCD, and summarizes some potential pharmacological agents for MASH treatment which exert their therapeutic effects partly via the regulation of hepatocyte PCD. These findings indicate that hepatocyte PCD putatively represents a new therapeutic point of intervention for MASH.
Collapse
Affiliation(s)
- Zilu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Rong Lin
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
15
|
Du K, Wang L, Jun JH, Dutta RK, Maeso-Díaz R, Oh SH, Ko DC, Diehl AM. Aging promotes metabolic dysfunction-associated steatotic liver disease by inducing ferroptotic stress. NATURE AGING 2024; 4:949-968. [PMID: 38918603 DOI: 10.1038/s43587-024-00652-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/17/2024] [Indexed: 06/27/2024]
Abstract
Susceptibility to the biological consequences of aging varies among organs and individuals. We analyzed hepatocyte transcriptomes of healthy young and aged male mice to generate an aging hepatocyte gene signature, used it to deconvolute transcriptomic data from humans and mice with metabolic dysfunction-associated liver disease, validated findings with functional studies in mice and applied the signature to transcriptomic data from other organs to determine whether aging-sensitive degenerative mechanisms are conserved. We discovered that the signature enriches in diseased livers in parallel with degeneration. It is also enriched in failing human hearts, diseased kidneys and pancreatic islets from individuals with diabetes. The signature includes genes that control ferroptosis. Aged mice develop more hepatocyte ferroptosis and liver degeneration than young mice when fed diets that induce metabolic stress. Inhibiting ferroptosis shifts the liver transcriptome of old mice toward that of young mice and reverses aging-exacerbated liver damage, identifying ferroptosis as a tractable, conserved mechanism for aging-related tissue degeneration.
Collapse
Affiliation(s)
- Kuo Du
- Department of Medicine, Duke University, Durham, NC, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Ji Hye Jun
- Department of Medicine, Duke University, Durham, NC, USA
| | - Rajesh K Dutta
- Department of Medicine, Duke University, Durham, NC, USA
| | | | - Seh Hoon Oh
- Department of Medicine, Duke University, Durham, NC, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
16
|
Venn-Watson S. The Cellular Stability Hypothesis: Evidence of Ferroptosis and Accelerated Aging-Associated Diseases as Newly Identified Nutritional Pentadecanoic Acid (C15:0) Deficiency Syndrome. Metabolites 2024; 14:355. [PMID: 39057678 PMCID: PMC11279173 DOI: 10.3390/metabo14070355] [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: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Ferroptosis is a newly discovered form of cell death caused by the peroxidation of fragile fatty acids in cell membranes, which combines with iron to increase reactive oxygen species and disable mitochondria. Ferroptosis has been linked to aging-related conditions, including type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease (NAFLD). Pentadecanoic acid (C15:0), an odd-chain saturated fat, is an essential fatty acid with the primary roles of stabilizing cell membranes and repairing mitochondrial function. By doing so, C15:0 reverses the underpinnings of ferroptosis. Under the proposed "Cellular Stability Hypothesis", evidence is provided to show that cell membranes optimally need >0.4% to 0.64% C15:0 to support long-term health and longevity. A pathophysiology of a newly identified nutritional C15:0 deficiency syndrome ("Cellular Fragility Syndrome") is provided that demonstrates how C15:0 deficiencies (≤0.2% total circulating fatty acids) can increase susceptibilities to ferroptosis, dysmetabolic iron overload syndrome, type 2 diabetes, cardiovascular disease, and NAFLD. Further, evidence is provided that C15:0 supplementation can reverse the described C15:0 deficiency syndrome, including the key components of ferroptosis. Given the declining dietary intake of C15:0, especially among younger generations, there is a need for extensive studies to understand the potential breadth of Cellular Fragility Syndrome across populations.
Collapse
Affiliation(s)
- Stephanie Venn-Watson
- Seraphina Therapeutics Inc., San Diego, CA 92106, USA;
- Epitracker Inc., San Diego, CA 92106, USA
| |
Collapse
|
17
|
Shen X, Yu Z, Wei C, Hu C, Chen J. Iron metabolism and ferroptosis in nonalcoholic fatty liver disease: what is our next step? Am J Physiol Endocrinol Metab 2024; 326:E767-E775. [PMID: 38506752 PMCID: PMC11376490 DOI: 10.1152/ajpendo.00260.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/06/2024] [Accepted: 03/09/2024] [Indexed: 03/21/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease with increasing prevalence worldwide. NAFLD could develop from simple hepatic steatosis to nonalcoholic steatohepatitis (NASH), NASH-related fibrosis, cirrhosis, and even hepatocellular carcinoma. However, the mechanism of NAFLD development has not yet been fully defined. Recently, emerging evidence shows that the dysregulated iron metabolism marked by elevated serum ferritin, and ferroptosis are involved in the NAFLD. Understanding iron metabolism and ferroptosis can shed light on the mechanisms of NAFLD development. Here, we summarized studies on iron metabolism and the ferroptosis process involved in NAFLD development to highlight potential medications and therapies for treating NAFLD.
Collapse
Affiliation(s)
- Xiang Shen
- Munich Medical Research School, Ludwig Maximilian University of Munich, Munich, Germany
| | - Ziqi Yu
- Munich Medical Research School, Ludwig Maximilian University of Munich, Munich, Germany
| | - Changli Wei
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Nanchang, People's Republic of China
| | - Chong Hu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Nanchang, People's Republic of China
| | - Jianyong Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Nanchang, People's Republic of China
| |
Collapse
|
18
|
Kim JY, Yang AY, Kim K, Kwon HH, Leem J, Kim YA. Pharmacological inhibition of p300 ameliorates steatosis, inflammation, and fibrosis in mice with non-alcoholic steatohepatitis. Heliyon 2024; 10:e30908. [PMID: 38774067 PMCID: PMC11107220 DOI: 10.1016/j.heliyon.2024.e30908] [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: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/24/2024] Open
Abstract
The histone acetyltransferase p300 plays a pivotal role in regulating gene expression and cellular phenotype through epigenetic mechanisms. It significantly influences lipid metabolism, which is a key factor in the pathogenesis of non-alcoholic steatohepatitis (NASH), by modulating the transcription of genes involved in lipid synthesis and accumulation. This study aimed to investigate the protective potential of inhibiting p300 in NASH. Male C57BL/6J mice were subjected to a methionine- and choline-deficient (MCD) diet for 4 weeks to induce NASH, and during this period, the p300 inhibitor C646 (10 mg/kg) was administered three times a week. C646 treatment reduced the elevation of p300 expression and histone H3 acetylation, leading to a decrease in liver injury markers in the serum and an improvement in the histological abnormalities observed in MCD diet-fed mice. C646 also reduced lipid accumulation by modulating de novo lipogenesis and suppressed inflammation, including cytokine overproduction and macrophage infiltration. Furthermore, C646 mitigated liver fibrosis and myofibroblast accumulation. This protective effect was achieved through the inhibition of apoptosis by reducing p53 and Bax expression and the suppression of ferroptosis by decreasing lipid peroxidation while enhancing antioxidant defenses. Additionally, C646 alleviated endoplasmic reticulum stress, as evidenced by the downregulation of unfolded protein response signaling molecules. These results highlight the potential of p300 as a therapeutic target for NASH.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Ah Young Yang
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Kiryeong Kim
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Hyun Hee Kwon
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Yun-A Kim
- Department of Family Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| |
Collapse
|
19
|
Pang S, Han B, Wu P, Yang X, Liu Y, Li J, Lv Z, Zhang Z. Resveratrol alleviates inorganic arsenic-induced ferroptosis in chicken brain via activation of the Nrf2 signaling pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 201:105885. [PMID: 38685251 DOI: 10.1016/j.pestbp.2024.105885] [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: 02/23/2024] [Revised: 03/09/2024] [Accepted: 03/23/2024] [Indexed: 05/02/2024]
Abstract
Inorganic arsenic (iAs) is a well-recognized environmental pollutant that induces severe brain injury in humans and animals. The antioxidant, anti-inflammatory, and anti-ferroptotic effects of resveratrol (Res) were demonstrated in multiple animal experiments. In order to investigate the protective effect of Res on iAs-induced chicken brain injury, the 40 chickens (19-d-old, female) brain injury model was established by oral administration of iAs (30 mg/L NaAsO2) for 6 weeks. All chickens had free access to both food and water during the experiment. The biochemical indices, hematoxylin-eosin staining, and related protein levels of oxidative stress, inflammation and ferroptosis were then determined. Our results indicated that Res (1000 mg/kg) alleviated the iAs-induced brain injury after 6 weeks of oral administration, primarily by reducing the interleukin-1β mRNA expression and nuclear factor kappa B and malondialdehyde level, and increasing the antioxidant enzyme activity and the mRNA expression of nuclear factor erythroid 2-related factor 2 (Nrf2). Taken together, our study demonstrates that Res effectively inhibits iAs-induced oxidative stress and ferroptosis by mediating the Nrf2 signaling pathway, thereby alleviating iAs-induced brain injury in chickens. This is the first time that the amelioration effects of Res on the iAs-induced brain have been investigated from multiple perspectives.
Collapse
Affiliation(s)
- Shan Pang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Biqi Han
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Xu Yang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Yunfeng Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Zhanjun Lv
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China.
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China.
| |
Collapse
|
20
|
Yang Z, He Y, Wu D, Shi W, Liu P, Tan J, Wang R, Yu B. Antiferroptosis therapy alleviated the development of atherosclerosis. MedComm (Beijing) 2024; 5:e520. [PMID: 38576455 PMCID: PMC10993356 DOI: 10.1002/mco2.520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/18/2024] [Accepted: 03/02/2024] [Indexed: 04/06/2024] Open
Abstract
Ferroptosis has been confirmed to be associated with various diseases, but the relationship between ferroptosis and atherosclerosis (AS) remains unclear. Our research detailly clarified the roles of ferroptosis in three continuous and main pathological stages of AS respectively (injury of endothelial cells [ECs], adhesion of monocytes, and formation of foam cells). We confirmed that oxidized low-density lipoprotein (ox-LDL), the key factor in the pathogenesis of AS, strongly induced ferroptosis in ECs. Inhibition of ferroptosis repressed the adhesion of monocytes to ECs by inhibiting inflammation of ECs. Ferroptosis also participated in the formation of foam cells and lipids by regulating the cholesterol efflux of macrophages. Further research confirmed that ox-LDL repressedthe activity of glutathione peroxidase 4 (GPX4), the classic lipid peroxide scavenger. Treatment of a high-fat diet significantly induced ferroptosis in murine aortas and aortic sinuses, which was accompanied by AS lesions and hyperlipidemia. Treatment with ferroptosis inhibitors significantly reduced ferroptosis, hyperlipidemia, and AS lesion development. In conclusion, our research determined that ox-LDL induced ferroptosis by repressing the activity of GPX4. Antiferroptosis treatment showed promising treatment effects in vivo. Ferroptosis-associated indexes also showed promising diagnostic potential in AS patients.
Collapse
Affiliation(s)
- Zhou Yang
- Department of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghaiChina
- Fudan Zhangjiang Institute, Fudan UniversityShanghaiChina
- Shanghai Key Laboratory of Vascular Lesions Regulation and RemodelingShanghaiChina
- Department of Head and Neck SurgeryFudan University Shanghai Cancer CenterShanghaiChina
| | - Yue He
- Shanghai University of Traditional Chinese MedicineShanghaiChina
- Department of CardiologyShanghai Eighth People's HospitalShanghaiChina
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Dejun Wu
- Department of General SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghaiChina
| | - Weihao Shi
- Department of Vascular SurgeryHuashan Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Ping Liu
- Shanghai University of Traditional Chinese MedicineShanghaiChina
- Shanghai University of Traditional Chinese MedicineDepartment of CardiologyLonghua HospitalShanghaiChina
| | - Jinyun Tan
- Department of Vascular SurgeryHuashan Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Rui Wang
- Department of Cardiovascular SurgeryNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Bo Yu
- Department of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghaiChina
- Fudan Zhangjiang Institute, Fudan UniversityShanghaiChina
- Shanghai Key Laboratory of Vascular Lesions Regulation and RemodelingShanghaiChina
- Department of Vascular SurgeryHuashan Hospital Affiliated to Fudan UniversityShanghaiChina
| |
Collapse
|
21
|
Wu IT, Yeh WJ, Huang WC, Yang HY. Very low-carbohydrate diet with higher protein ratio improves lipid metabolism and inflammation in rats with diet-induced nonalcoholic fatty liver disease. J Nutr Biochem 2024; 126:109583. [PMID: 38244701 DOI: 10.1016/j.jnutbio.2024.109583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/27/2023] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is commonly associated with obesity, and it is mainly treated through lifestyle modifications. The very low-carbohydrate diet (VLCD) can help lose weight rapidly but the possible effects of extreme dietary patterns on lipid metabolism and inflammatory responses in individuals with NAFLD remain debatable. Moreover, VLCD protein content may affect its effectiveness in weight loss, steatosis, and inflammatory responses. Therefore, we investigated the effects of VLCDs with different protein contents in NAFLD rats and the mechanisms underlying these effects. After a 16-week inducing period, the rats received an isocaloric normal diet (NC group) or a VLCD with high or low protein content (NVLH vs. NVLL group, energy ratio:protein/carbohydrate/lipid=20/1/79 vs. 6/1/93) for the next 8 weeks experimental period. We noted that the body weight decreased in both the NVLH and NVLL groups; nevertheless, the NVLH group demonstrated improvements in ketosis. The NVLL group led to hepatic lipid accumulation, possibly by increasing very-low-density lipoprotein receptor (VLDLR) expression and elevating liver oxidative stress, subsequently activating the expression of Nrf2, and inflammation through the TLR4/TRIF/NLRP3 and TLR4/MyD88/NF-κB pathway. The NVLH was noted to prevent the changes in VLDLR and the TLR4-inflammasome pathway partially. The VLCD also reduced the diversity of gut microbiota and changed their composition. In conclusion, although low-protein VLCD consumption reduces BW, it may also lead to metabolic disorders and changes in microbiota composition; nevertheless, a VLCD with high protein content may partially alleviate these limitations.
Collapse
Affiliation(s)
- I-Ting Wu
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wan-Ju Yeh
- Graduate Program of Nutrition Science, National Taiwan Normal University, Taipei, Taiwan
| | - Wen-Chih Huang
- Department of Anatomical Pathology, Taipei Institute of Pathology, Taipei City, Taiwan
| | - Hsin-Yi Yang
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
22
|
Torshin IY, Gromova OA, Bogacheva TE. Systematic analysis of the relationship between non-alcoholic fatty liver disease and tissue iron overload: promising areas for the use of polypeptide therapy. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2024:139-152. [DOI: 10.31146/1682-8658-ecg-218-10-139-152] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Iron overload in non-alcoholic fatty liver disease (NAFLD) is a fairly common phenomenon that receives very little attention in clinical practice. However, iron overload, leading to hemosiderosis (deposition of “indigestible” nanodispersed iron oxides in various tissues) significantly aggravates NAFLD, stimulating increased chronic inflammation, insulin resistance and hemosiderosis of other organs. As a result, ferroptosis of hepatocytes occurs (apoptosis caused by iron overload and hemosiderosis), which accelerates the transformation of non-alcoholic steatosis into non-alcoholic steatohepatitis (NASH) and, subsequently, into liver cirrhosis. Iron overload is aggravated by micronutrient deficiencies and pathogenic intestinal microbiota. The paper presents the results of a systematic analysis of this issue, describes the prospects for therapy using micronutrients and human placenta hydrolysates (HPP), which contribute not only to the regeneration of liver tissue, but also to the normalization of iron homeostasis.
Collapse
Affiliation(s)
- I. Yu. Torshin
- Federal Research Center “Computer Science and Control” of Russian Academy of Sciences
| | - O. A. Gromova
- Federal Research Center “Computer Science and Control” of Russian Academy of Sciences
| | | |
Collapse
|
23
|
Xia Q, Lan J, Pan Y, Wang Y, Song T, Yang Y, Tian X, Chen L, Gu Z, Ding YY. Effects of Dityrosine on Lactic Acid Metabolism in Mice Gastrocnemius Muscle During Endurance Exercise via the Oxidative Stress-Induced Mitochondria Damage. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:5269-5282. [PMID: 38439706 DOI: 10.1021/acs.jafc.3c09649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Dityrosine (Dityr) has been detected in commercial food as a product of protein oxidation and has been shown to pose a threat to human health. This study aims to investigate whether Dityr causes a decrease in lactic acid metabolism in the gastrocnemius muscle during endurance exercise. C57BL/6 mice were administered Dityr or saline by gavage for 13 weeks and underwent an endurance exercise test on a treadmill. Dityr caused a severe reduction in motion displacement and endurance time, along with a significant increase in lactic acid accumulation in the blood and gastrocnemius muscle in mice after exercise. Dityr induced significant mitochondrial defects in the gastrocnemius muscle of mice. Additionally, Dityr induced serious oxidative stress in the gastrocnemius muscle, accompanied by inflammation, which might be one of the causes of mitochondrial dysfunction. Moreover, significant apoptosis in the gastrocnemius muscle increased after exposure to Dityr. This study confirmed that Dityr induced oxidative stress in the gastrocnemius muscle, which further caused significant mitochondrial damage in the gastrocnemius muscle cell, resulting in decreased capacity of lactic acid metabolism and finally affected performance in endurance exercise. This may be one of the possible mechanisms by which highly oxidized foods cause a decreased muscle energy metabolism.
Collapse
Affiliation(s)
- Qiudong Xia
- Department of Physical Education, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Jinchi Lan
- Food Safety Key Laboratory of Zhejiang Province, National Experimental Teaching Demonstration Center for Food Engineering and Quality and Safety, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Yuxiang Pan
- Food Safety Key Laboratory of Zhejiang Province, National Experimental Teaching Demonstration Center for Food Engineering and Quality and Safety, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Yuxin Wang
- Food Safety Key Laboratory of Zhejiang Province, National Experimental Teaching Demonstration Center for Food Engineering and Quality and Safety, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Tianyuan Song
- Food Safety Key Laboratory of Zhejiang Province, National Experimental Teaching Demonstration Center for Food Engineering and Quality and Safety, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Ying Yang
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xu Tian
- Beijing Competitor Sports Nutrition Research Institute, Beijing 100027, China
| | - Longjun Chen
- Huzhou Shengtao Biotechnology LLC, Huzhou 313000, China
| | - Zhenyu Gu
- Food Safety Key Laboratory of Zhejiang Province, National Experimental Teaching Demonstration Center for Food Engineering and Quality and Safety, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Yin-Yi Ding
- Food Safety Key Laboratory of Zhejiang Province, National Experimental Teaching Demonstration Center for Food Engineering and Quality and Safety, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| |
Collapse
|
24
|
Yuan M, Wang Y, Tian X, Zheng W, Zuo H, Zhang X, Song H. Ferrostatin-1 improves prognosis and regulates gut microbiota of steatotic liver transplantation recipients in rats. Future Microbiol 2024; 19:413-429. [PMID: 38305222 DOI: 10.2217/fmb-2023-0133] [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: 06/09/2023] [Accepted: 11/15/2023] [Indexed: 02/03/2024] Open
Abstract
Aims: To investigate the effects of Ferrostatin-1 (Fer-1) on improving the prognosis of liver transplant recipients with steatotic liver grafts and regulating gut microbiota in rats. Methods: We obtained steatotic liver grafts and established a liver transplantation model. Recipients were divided into sham, liver transplantation and Fer-1 treatment groups, which were assessed 1 and 7 days after surgery (n = 6). Results & conclusion: Fer-1 promotes recovery of the histological structure and function of steatotic liver grafts and the intestinal tract, and improves inflammatory responses of recipients following liver transplantation. Fer-1 reduces gut microbiota pathogenicity, and lowers iron absorption and improves fat metabolism of recipients, thereby protecting steatotic liver grafts.
Collapse
Affiliation(s)
- Mengshu Yuan
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Yuxin Wang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Xiaorong Tian
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Weiping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, 300192, PR China
- NHC Key Laboratory of Critical Care Medicine, Tianjin, 300192, PR China
| | - Huaiwen Zuo
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Xinru Zhang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Hongli Song
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, 300192, PR China
- Tianjin Key Laboratory of Organ Transplantation, Tianjin, PR China
| |
Collapse
|
25
|
Theys C, Vanderhaeghen T, Van Dijck E, Peleman C, Scheepers A, Ibrahim J, Mateiu L, Timmermans S, Vanden Berghe T, Francque SM, Van Hul W, Libert C, Vanden Berghe W. Loss of PPARα function promotes epigenetic dysregulation of lipid homeostasis driving ferroptosis and pyroptosis lipotoxicity in metabolic dysfunction associated Steatotic liver disease (MASLD). FRONTIERS IN MOLECULAR MEDICINE 2024; 3:1283170. [PMID: 39086681 PMCID: PMC11285560 DOI: 10.3389/fmmed.2023.1283170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/14/2023] [Indexed: 08/02/2024]
Abstract
Metabolic Dysfunction Associated Steatotic Liver Disease (MASLD) is a growing epidemic with an estimated prevalence of 20%-30% in Europe and the most common cause of chronic liver disease worldwide. The onset and progression of MASLD are orchestrated by an interplay of the metabolic environment with genetic and epigenetic factors. Emerging evidence suggests altered DNA methylation pattern as a major determinant of MASLD pathogenesis coinciding with progressive DNA hypermethylation and gene silencing of the liver-specific nuclear receptor PPARα, a key regulator of lipid metabolism. To investigate how PPARα loss of function contributes to epigenetic dysregulation in MASLD pathology, we studied DNA methylation changes in liver biopsies of WT and hepatocyte-specific PPARα KO mice, following a 6-week CDAHFD (choline-deficient, L-amino acid-defined, high-fat diet) or chow diet. Interestingly, genetic loss of PPARα function in hepatocyte-specific KO mice could be phenocopied by a 6-week CDAHFD diet in WT mice which promotes epigenetic silencing of PPARα function via DNA hypermethylation, similar to MASLD pathology. Remarkably, genetic and lipid diet-induced loss of PPARα function triggers compensatory activation of multiple lipid sensing transcription factors and epigenetic writer-eraser-reader proteins, which promotes the epigenetic transition from lipid metabolic stress towards ferroptosis and pyroptosis lipid hepatoxicity pathways associated with advanced MASLD. In conclusion, we show that PPARα function is essential to support lipid homeostasis and to suppress the epigenetic progression of ferroptosis-pyroptosis lipid damage associated pathways towards MASLD fibrosis.
Collapse
Affiliation(s)
- Claudia Theys
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tineke Vanderhaeghen
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Cedric Peleman
- Laboratory of Experimental Medicine and Pediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Pathophysiology Lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anne Scheepers
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Joe Ibrahim
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Ligia Mateiu
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Steven Timmermans
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Pathophysiology Lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sven M. Francque
- Laboratory of Experimental Medicine and Pediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Wim Van Hul
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wim Vanden Berghe
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
26
|
Bu G, Chen G, Li J, Wu D, Liao J. Bifidobacterium bifidum BGN4 fractions ameliorate palmitic acid-induced hepatocyte ferroptosis by inhibiting SREBP1-CYP2E1 pathway. J Investig Med 2024; 72:67-79. [PMID: 37723650 DOI: 10.1177/10815589231204058] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is strongly associated with disturbances in the intestinal microbiota. Herein, the biological effects and mechanism of Bifidobacterium bifidum BGN4 fractions in regulating hepatocyte ferroptosis during MAFLD progression were investigated. To establish an in vitro model of MAFLD, LO2 cells were subjected to palmitic acid (PA). The mRNA and protein expressions were assessed using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot, respectively. LO2 cell proliferation was examined using 5-diphenyltetrazolium bromide (MTT) and ethynyl-2'-deoxyuridine (EdU) assays, whereas its apoptosis was evaluated by flow cytometry. Furthermore, level of reactive oxygen species (ROS) was measured using 2', 7,-Dichlorodihydrofluorescein diacetate (DCFH-DA) staining. Additionally, the levels of Fe2+, malondialdehyde (MDA), and glutathione (GSH), as well as the activities of superoxide dismutase (SOD) and glutathione peroxidase (GPX) were detected using corresponding kits. Chromatin immunoprecipitation and dual-luciferase reporter gene assays were performed to analyze the interaction between sterol-regulatory element binding protein 1 (SREBP1) and cytochrome P450-2E1 (CYP2E1) promoter. Our results revealed that Bifidobacterium bifidum BGN4 fractions effectively ameliorated PA-induced hepatocyte injury, oxidative stress, and ferroptosis. However, these beneficial effects of BGN4 fractions on PA-induced hepatocyte were dramatically reversed by SREBP1 overexpression, suggesting that BGN4 attenuated MAFLD by acting on SREBP1. Moreover, we observed that BGN4 fractions inhibited CYP2E1 transcription by suppressing SREBP1 nuclear translocation. In addition, CYP2E1 overexpression eliminated the inhibitory effect of BGN4 fractions on PA-induced hepatocyte oxidative stress and ferroptosis. These findings collectively indicated that BGN4 fractions reduced CYP2E1 expression by inhibiting SREBP1 nuclear translocation, thereby suppressing hepatocyte oxidative stress and ferroptosis during the development of MAFLD.
Collapse
Affiliation(s)
- Guangkui Bu
- Department of Gastroenterology, Hunan Provincial People's Hospital The First-Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Gang Chen
- Department of Gastroenterology, Hunan Provincial People's Hospital The First-Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Juan Li
- Department of Gastroenterology, Hunan Provincial People's Hospital The First-Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Dan Wu
- Department of Gastroenterology, Hunan Provincial People's Hospital The First-Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Jiangtao Liao
- Department of Gastroenterology, Hunan Provincial People's Hospital The First-Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| |
Collapse
|
27
|
Afarin R, Dinarvand N, Jaberian Asl B, Orak G, Shakerian E, Bineshfar F, Ahangarpour A. Comparison of the anti-inflammatory and antilipidemic activity of diosmin and saroglitazar in a model of nonalcoholic fatty liver induced by a high-fat diet in Wistar rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:241-246. [PMID: 38234671 PMCID: PMC10790293 DOI: 10.22038/ijbms.2023.70157.15255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 09/27/2023] [Indexed: 01/19/2024]
Abstract
Objectives Non-alcoholic fatty liver disease (NAFLD) is the most common liver-related metabolic disorder in the world, with a global prevalence of 25%. Compounds with anti-inflammatory, lipid-lowering, and insulin-sensitizing properties can be used for the prevention or treatment of NAFLD. Therefore, this study was conducted to investigate the effect of saroglitazar (a dual PPARα/γ agonist) and diosmin (a flavonoid) on non-alcoholic fatty liver induced by a high-fat diet (HFD) in Wistar rats. Materials and Methods Forty male Wistar rats (6-8 weeks old) were fed an HFD to induce NAFLD. After 7 weeks, rats were divided into four groups: group1 was fed HFD, and the other groups received HFD+saroglitazar, HFD+diosmin, and HFD+ saroglitazar+diosmin. We examined body and liver weight, histopathology, serum levels of liver enzymes (ALT and AST), and lipid profiles (LDL-C and HDL-C) using the standard protocols. qRT-PCR was also used to examine the expression of PPARα, PPARγ, SREBP1c, FAS, ACC, CPT1α, and pro-inflammatory genes (IL6, TNFα, and TGFβ). Results Rats fed the HFD showed characteristics of NAFLD (pathologically and biochemically). Administration of saroglitazar and diosmin alone caused a significant decrease in the levels of PPARγ, SREBP1c, FAS, ACC, ALT, AST, LDL-C, and pro-inflammatory genes and a significant increase in PPARα, CPT1a, and HDL-C in comparison with the HF group (P<0.05). Their combined effect was more evident. Conclusion Our results showed that diosmin, like saroglitazar, significantly ameliorated inflammatory and lipid profiles in HFD-induced NAFLD, suggesting that diosmin, as a natural compound, could be a suitable alternative to saroglitazar.
Collapse
Affiliation(s)
- Reza Afarin
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Negar Dinarvand
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahar Jaberian Asl
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ghazal Orak
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elham Shakerian
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Bineshfar
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Akram Ahangarpour
- Diabetes Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
28
|
Qin J, Cao P, Ding X, Zeng Z, Deng L, Luo L. Machine learning identifies ferroptosis-related gene ANXA2 as potential diagnostic biomarkers for NAFLD. Front Endocrinol (Lausanne) 2023; 14:1303426. [PMID: 38192427 PMCID: PMC10773757 DOI: 10.3389/fendo.2023.1303426] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/14/2023] [Indexed: 01/10/2024] Open
Abstract
Introduction Non-alcoholic fatty liver disease (NAFLD), a major cause of chronic liver disease, still lacks effective therapeutic targets today. Ferroptosis, a type of cell death characterized by lipid peroxidation, has been linked to NAFLD in certain preclinical trials, yet the exact molecular mechanism remains unclear. Thus, we analyzed the relationship between ferroptosis genes and NAFLD using high-throughput data. Method We utilized a total of 282 samples from five datasets, including two mouse ones, one human one, one single nucleus dataset and one single cell dataset from Gene Expression Omnibus (GEO), as the data basis of our study. To filter robust treatment targets, we employed four machine learning methods (LASSO, SVM, RF and Boruta). In addition, we used an unsupervised consensus clustering algorithm to establish a typing scheme for NAFLD based on the expression of ferroptosis related genes (FRGs). Our study is also the first to investigate the dynamics of FRGs throughout the disease process by time series analysis. Finally, we validated the relationship between core gene and ferroptosis by in vitro experiments on HepG2 cells. Results We discovered ANXA2 as a central focus in NAFLD and indicated its potential to boost ferroptosis in HepG2 cells. Additionally, based on the results obtained from time series analysis, ANXA2 was observed to significantly define the disease course of NAFLD. Our results demonstrate that implementing a ferroptosis-based staging method may hold promise for the diagnosis and treatment of NAFLD. Conclusion Our findings suggest that ANXA2 may be a useful biomarker for the diagnosis and characterization of NAFLD.
Collapse
Affiliation(s)
- Jingtong Qin
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Peng Cao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuexuan Ding
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Zeyao Zeng
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Liyan Deng
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| |
Collapse
|
29
|
Zhang H, Axinbai M, Zhao Y, Wei J, Qu T, Kong J, He Y, Zhang L. Bioinformatics analysis of ferroptosis-related genes and immune cell infiltration in non-alcoholic fatty liver disease. Eur J Med Res 2023; 28:605. [PMID: 38115130 PMCID: PMC10729346 DOI: 10.1186/s40001-023-01457-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 10/18/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND The morbidity and mortality rates of patients with non-alcoholic fatty liver disease (NAFLD) have been steadily increasing in recent years. Previous studies have confirmed the important role of ferroptosis in NAFLD development; however, the precise mechanism through which ferroptosis influences NAFLD occurrence remains unclear. The present study aimed to identify and validate ferroptosis-related genes involved in NAFLD pathogenesis and to investigate the underlying molecular mechanisms of NAFLD. METHODS We downloaded microarray datasets GSE72756 and GSE24807 to identify differentially expressed genes (DEGs) between samples from healthy individuals and patients with NAFLD. From these DEGs, we extracted ferroptosis-related DEGs. GSE89632, another microarray dataset, was used to validate the expression of ferroptosis-related genes. A protein-protein interaction (PPI) network of ferroptosis-related genes was then constructed. The target genes were also subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Finally, competing endogenous RNA networks were constructed. We used the CIBERSORT package to evaluate the infiltration of immune cells infiltration in NAFLD. RESULTS Five ferroptosis-related genes (SCP2, MUC1, DPP4, SLC1A4, and TF) were identified as promising diagnostic biomarkers for NAFLD. Enrichment analyses revealed that these genes are mainly involved in metabolic processes. NEAT1-miR-1224-5p-SCP2, NEAT1-miR-485-5p-MUC1, MALAT1-miR-485-5p-MUC1, and CNOT6-miR-145-5p-SLC1A4 are likely to be the potential RNA regulatory pathways that affect NAFLD development. Principal component analysis indicated significant differences in immune cell infiltration between the two groups. CONCLUSIONS This study identified five ferroptosis-related genes as potential biomarkers for diagnosing NAFLD. The correlations between the expression of ferroptosis-related genes and immune cell infiltration might shed light on the study of the molecular mechanism underlying NAFLD development.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Digestion, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Malina Axinbai
- Department of Digestion, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
- Xinjiang Medical University, Urumqi, China
| | - Yuqing Zhao
- Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaoyang Wei
- Department of Digestion, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Tongshuo Qu
- Department of Digestion, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Jingmin Kong
- Department of Emergency, Beijing Chaoyang Integrative Medicine Rescue and First Aid Hospital, Beijing, China
| | - Yongqiang He
- Department of Digestion, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Liping Zhang
- Department of Digestion, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
- Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
30
|
Jiang T, Xiao Y, Zhou J, Luo Z, Yu L, Liao Q, Liu S, Qi X, Zhang H, Hou M, Miao W, Batsaikhan B, Damba T, Liang Y, Li Y, Zhou L. Arbutin alleviates fatty liver by inhibiting ferroptosis via FTO/SLC7A11 pathway. Redox Biol 2023; 68:102963. [PMID: 37984229 PMCID: PMC10694775 DOI: 10.1016/j.redox.2023.102963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/26/2023] [Accepted: 11/10/2023] [Indexed: 11/22/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a potentially serious disease that affects 30 % of the global population and poses a significant risk to human health. However, to date, no safe, effective and appropriate treatment modalities are available. In recent years, ferroptosis has emerged as a significant mode of cell death and has been found to play a key regulatory role in the development of NAFLD. In this study, we found that arbutin (ARB), a natural antioxidant derived from Arctostaphylos uva-ursi (L.), inhibits the onset of ferroptosis and ameliorates high-fat diet-induced NAFLD in vivo and in vitro. Using reverse docking, we identified the demethylase fat mass and obesity-related protein (FTO) as a potential target of ARB. Subsequent mechanistic studies revealed that ARB plays a role in controlling methylation of the SLC7A11 gene through inhibition of FTO. In addition, we demonstrated that SLC7A11 could alleviate the development of NAFLD in vivo and in vitro. Our findings identify the FTO/SLC7A11 axis as a potential therapeutic target for the treatment of NAFLD. Specifically, we show that ARB alleviates NAFLD by acting on the FTO/SLC7A11 pathway to inhibit ferroptosis.
Collapse
Affiliation(s)
- Tianyu Jiang
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Yao Xiao
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Jinfeng Zhou
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Zupeng Luo
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Lin Yu
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Qichao Liao
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Siqi Liu
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Xinyi Qi
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Hao Zhang
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Menglong Hou
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - WeiWei Miao
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Batbold Batsaikhan
- Department of Internal Medicine, Institute of Medical Sciences, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia; Department of Health Research, Graduate School, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Turtushikh Damba
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Yunxiao Liang
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Yixing Li
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China.
| | - Lei Zhou
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| |
Collapse
|
31
|
Jin X, He R, Lin Y, Liu J, Wang Y, Li Z, Liao Y, Yang S. Shenshuaifu Granule Attenuates Acute Kidney Injury by Inhibiting Ferroptosis Mediated by p53/SLC7A11/GPX4 Pathway. Drug Des Devel Ther 2023; 17:3363-3383. [PMID: 38024532 PMCID: PMC10656853 DOI: 10.2147/dddt.s433994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Acute kidney injury (AKI) is a common clinical condition resulting in a rapid decline in renal function, and requires improvement in effective preventive measures. Ferroptosis, a novel form of cell death, is closely related to AKI. Shenshuaifu granule (SSF) has been demonstrated to prevent AKI through suppressing inflammation and apoptosis. Objective This study aimed to explore whether SSF can inhibit ferroptosis in AKI. Methods Active ingredients in SSF were detected through HPLC-MS/MS, and their binding abilities with ferroptosis were evaluated by molecular docking. Then, male C57/BL/6J mice were randomly divided into control, cisplatin, and cisplatin+SSF groups. In the latter two groups, mice were intraperitoneally injected with 20 mg/kg of cisplatin. For five consecutive days prior to cisplatin injection, mice in the cisplatin+SSF group were gavaged with 5.2 g/kg of SSF per day.72 h after cisplatin injection, the mice were sacrificed. Serum creatinine (SCr) and blood urea nitrogen (BUN) were measured to evaluate renal function. H&E and PAS staining were used to observe pathological damage of kidney. Cell death was observed by TUNEL staining, and iron accumulation in kidneys of mice was detected by Prussian blue staining. Western blotting, immunohistochemistry, and immunofluorescence were used to investigate the presence of inflammation, oxidative stress, mitochondrial dysfunction, iron deposition, and lipid peroxidation in mouse kidneys. Results Active ingredients in SSF had strong affinities with ferroptosis. SSF reduced SCr (p<0.01) and BUN (p<0.0001) levels, pathological damage (p<0.0001), dead cells in the tubular epithelium (p<0.0001) and iron deposition (p<0.01) in mice with cisplatin induced AKI. And SSF downregulated macrophage infiltration (p<0.01), the expressions of high mobility group box 1 (HMGB1, p<0.05) and interleukin (IL)-17 (p<0.05), upregulated superoxide dismutase (SOD) 1 and 2 (p<0.01), and catalase (CAT, p<0.05), and alleviated mitochondrial dysfunction (p<0.05). More importantly, SSF regulated iron transport and intracellular iron overload and reduced the expression of ferritin (p<0.05). Moreover, it downregulated the expressions of cyclo-oxygenase-2 (Cox-2, p<0.001), acid CoA ligase 4 (ACSL4, p<0.05), and solute carrier family 7, member 11 (SLC7A11, p<001), upregulated glutathione peroxidase 4 (GPX4, p<0.01) and p53 (p<0.01), and decreased 4-hydroxynonenal (4-HNE) level (p<0.001). Conclusion SSF attenuates AKI by inhibiting ferroptosis mediated by p53/SLC7A11/GPX4 pathway.
Collapse
Affiliation(s)
- Xiaoming Jin
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Riming He
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Yunxin Lin
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Jiahui Liu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Yuzhi Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Zhongtang Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Yijiao Liao
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Shudong Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, People’s Republic of China
| |
Collapse
|
32
|
Ramírez-Mejía MM, Qi X, Abenavoli L, Romero-Gómez M, Eslam M, Méndez-Sánchez N. Metabolic dysfunction: The silenced connection with fatty liver disease. Ann Hepatol 2023; 28:101138. [PMID: 37468095 DOI: 10.1016/j.aohep.2023.101138] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 07/21/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a global public health burden. Despite the increase in its prevalence, the disease has not received sufficient attention compared to the associated diseases such as diabetes mellitus and obesity. In 2020 it was proposed to rename NAFLD to metabolic dysfunction-associated fatty liver disease (MAFLD) in order to recognize the metabolic risk factors and the complex pathophysiological mechanisms associated with its development. Furthermore, along with the implementation of the proposed diagnostic criteria, the aim is to address the whole clinical spectrum of the disease, regardless of BMI and the presence of other hepatic comorbidities. As would it be expected with such a paradigm shift, differing viewpoints have emerged regarding the benefits and disadvantages of renaming fatty liver disease. The following review aims to describe the way to the MAFLD from a historical, pathophysiological and clinical perspective in order to highlight why MAFLD is the approach to follow.
Collapse
Affiliation(s)
- Mariana M Ramírez-Mejía
- Plan of Combined Studies in Medicine (PECEM-MD/PhD), Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico; Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Liaoning Province, China
| | - Ludovico Abenavoli
- Department of Health Sciences, University Magna Graecia of Catanzaro, Italy
| | - Manuel Romero-Gómez
- Digestive Diseases Unit, Department of Medicine, SeLiver Group, Institute of Biomedicine of Sevilla (HUVR/CSIC/US), University of Seville, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico; Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
33
|
Blas-García A, Apostolova N. Novel Therapeutic Approaches to Liver Fibrosis Based on Targeting Oxidative Stress. Antioxidants (Basel) 2023; 12:1567. [PMID: 37627562 PMCID: PMC10451738 DOI: 10.3390/antiox12081567] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Chronic liver disease (CLD) constitutes a growing global health issue, with no effective treatments currently available. Oxidative stress closely interacts with other cellular and molecular processes to trigger stress pathways in different hepatic cells and fuel the development of liver fibrosis. Therefore, inhibition of reactive oxygen species (ROS)-mediated effects and modulation of major antioxidant responses to counteract oxidative stress-induced damage have emerged as interesting targets to prevent or ameliorate liver injury. Although many preclinical studies have shown that dietary supplements with antioxidant properties can significantly prevent CLD progression in animal models, this strategy has not proved effective to significantly reduce fibrosis when translated into clinical trials. Novel and more specific therapeutic approaches are thus required to alleviate oxidative stress and reduce liver fibrosis. We have reviewed the relevant literature concerning the crucial role of alterations in redox homeostasis in different hepatic cell types during the progression of CLD and discussed current pharmacological approaches to ameliorate fibrosis by reducing oxidative stress focusing on selective modulation of enzymatic oxidant sources, antioxidant systems and ROS-mediated pathogenic processes.
Collapse
Affiliation(s)
- Ana Blas-García
- Departamento de Fisiología, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Av. de Catalunya, 21, 46020 Valencia, Spain
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Nadezda Apostolova
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Av. de Catalunya, 21, 46020 Valencia, Spain
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Departamento de Farmacología, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| |
Collapse
|
34
|
Yuan X, Li L, Zhang Y, Ai R, Li D, Dou Y, Hou M, Zhao D, Zhao S, Nan Y. Heme oxygenase 1 alleviates nonalcoholic steatohepatitis by suppressing hepatic ferroptosis. Lipids Health Dis 2023; 22:99. [PMID: 37422643 DOI: 10.1186/s12944-023-01855-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/22/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND Heme oxygenase 1 (HO-1) has an influential but insufficiently investigated effect on ferroptosis, which is a novel form of programmed cell death and may play an effect on nonalcoholic steatohepatitis (NASH). However, the understanding of the mechanism is limited. Herein, our study aimed to explore the mechanism and role of HO-1 in NASH ferroptosis. METHODS Hepatocyte conditional HO-1 knockout (HO-1HEPKO) C57BL/6J mice were established and fed a high-fat diet (HFD). Additionally, wild-type mice were fed either a normal diet or a HFD. Hepatic steatosis, inflammation, fibrosis, lipid peroxidation, and iron overload were assessed. AML12 and HepG2 cells were used to investigate the underlying mechanisms in vitro. Finally, liver sections from NASH patients were used to clinically validate the histopathology of ferroptosis. RESULTS In mice, HFD caused lipid accumulation, inflammation, fibrosis, and lipid peroxidation, which were aggravated by HO-1HEPKO. In line with the in vivo results, HO-1 knockdown upregulated reactive oxygen species accumulation, lipid peroxidation, and iron overload in AML12 and HepG2 cells. Additionally, HO-1 knockdown reduced the GSH and SOD levels, which was in contrast to HO-1 overexpression in vitro. Furthermore, the present study revealed that the NF-κB signaling pathway was associated with ferroptosis in NASH models. Likewise, these findings were consistent with the liver histopathology results of NASH patients. CONCLUSION The current study showed that HO-1 could alleviate NASH progression by mediating ferroptosis.
Collapse
Affiliation(s)
- Xiwei Yuan
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Lu Li
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Ying Zhang
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Rong Ai
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Dongdong Li
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Yao Dou
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Mengmeng Hou
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Dandan Zhao
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Suxian Zhao
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China.
| |
Collapse
|
35
|
Honma K, Kirihara S, Nakayama H, Fukuoka T, Ohara T, Kitamori K, Sato I, Hirohata S, Fujii M, Yamamoto S, Ran S, Watanabe S. Selective autophagy associated with iron overload aggravates non-alcoholic steatohepatitis via ferroptosis. Exp Biol Med (Maywood) 2023; 248:1112-1123. [PMID: 37646078 PMCID: PMC10583757 DOI: 10.1177/15353702231191197] [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: 02/01/2023] [Accepted: 05/18/2023] [Indexed: 09/01/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a progressive form of non-alcoholic fatty liver disease (NAFLD) that causes cirrhosis and hepatocellular carcinoma. Iron is an essential trace element in the body; however, excess iron can cause tissue damage and dysfunction. Iron overload is often observed in patients with NASH, and the amount of iron accumulated in the liver positively correlates with the histological severity of NASH. Ferroptosis, a novel form of iron-dependent cell death, is caused by the accumulation of lipid peroxidation and oxidative stress and is related to NASH. In addition, ferroptosis is closely related to autophagy, an intracellular self-degradation process. Although autophagy has many beneficial effects, it may also be harmful to the organism, for example, inducing ferroptosis. It is unclear whether iron overload aggravates NASH via autophagy. The aim of this research is to determine the mechanism by which iron overload induces ferroptosis via autophagy and aggravates NASH. Stroke-prone spontaneously hypertensive rats (SHRSP5/Dmcr) were divided into two groups and fed a high-fat and high-cholesterol (HFC) diet for eight weeks. Iron dextran was administered to the Fe group in addition to the HFC diet. Blood analysis, histological staining, calcineurin activity assay, quantitative reverse transcription polymerase chain reaction (RT-PCR), immunofluorescence staining, and electron microscopy were performed. The results showed that iron overload promoted autophagy via nuclear translocation of transcription factor EB (TFEB) and induced ferritinophagy, which is the autophagic degradation of ferritin. In addition, the HFC diet induced lipophagy, the autophagic degradation of lipid droplets. The Fe group also exhibited promoted ferroptosis and aggravated hepatic inflammation and fibrosis. In conclusion, iron overload accelerates ferritinophagy and lipophagy, aggravating NASH pathology via ferroptosis. These findings indicate the therapeutic potential of inhibiting autophagy and ferroptosis for treating NASH.
Collapse
Affiliation(s)
- Koki Honma
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan
| | - Sora Kirihara
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan
| | - Hinako Nakayama
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan
| | - Taketo Fukuoka
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan
| | - Toshiaki Ohara
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama University, Okayama 700-8558, Japan
| | - Kazuya Kitamori
- College of Human Life and Environment, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Ikumi Sato
- Academic Field of Health Science, Okayama University, Okayama 700-8558, Japan
| | - Satoshi Hirohata
- Academic Field of Health Science, Okayama University, Okayama 700-8558, Japan
| | - Moe Fujii
- Department of Medical Technology, Ehime Prefectural University of Health Sciences, Ehime 791-2101, Japan
| | - Shusei Yamamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan
- Academic Field of Health Science, Okayama University, Okayama 700-8558, Japan
| | - Shang Ran
- HeiLongjiang Provincial Center for disease control and prevention, Harbin 150030, China
| | - Shogo Watanabe
- Academic Field of Health Science, Okayama University, Okayama 700-8558, Japan
| |
Collapse
|
36
|
Kulkarni AV, Sarin SK. The bidirectional impacts of alcohol consumption and MAFLD for progressive fatty liver disease. Ther Adv Endocrinol Metab 2023; 14:20420188231178370. [PMID: 37323163 PMCID: PMC10265351 DOI: 10.1177/20420188231178370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), once considered a benign condition, has been associated with several cardiometabolic complications over the past two decades. The worldwide prevalence of NAFLD is as high as 30%. NAFLD requires the absence of a "significant alcohol intake." Conflicting reports have suggested that moderate alcohol consumption may be protective; therefore, the diagnosis of NAFLD previously relied on negative criteria. However, there has been a significant increase in alcohol consumption globally. Apart from the rise in alcohol-related liver disease (ARLD), alcohol, a major toxin, is associated with an increased risk of several cancers, including hepatocellular carcinoma. Alcohol misuse is a significant contributor to disability-adjusted life years. Recently, the term metabolic dysfunction-associated fatty liver disease (MAFLD) was proposed instead of NAFLD to include the metabolic dysfunction responsible for the major adverse outcomes in patients with fatty liver disease. MAFLD, dependent on the "positive diagnostic criteria" rather than previous exclusion criteria, may identify individuals with poor metabolic health and aid in managing patients at increased risk of all-cause and cardiovascular mortality. Although MAFLD is less stigmatizing than NAFLD, excluding alcohol intake may increase the risk of already existing underreported alcohol consumption in this subgroup of patients. Therefore, alcohol consumption may increase the prevalence of fatty liver disease and its associated complications in patients with MAFLD. This review discusses the effects of alcohol intake and MAFLD on fatty liver disease.
Collapse
|
37
|
Hu C, Zhao JF, Wang YM, Wu XL, Ye L. Tiliroside induces ferroptosis to repress the development of triple-negative breast cancer cells. Tissue Cell 2023; 83:102116. [PMID: 37301139 DOI: 10.1016/j.tice.2023.102116] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023]
Abstract
Ferroptosis is a newly found form of non-apoptotic regulated cell death that is essential for the advancement of cancer. Tiliroside (Til), an effective natural flavonoid glycoside of oriental paperbush flower, has been explored as a potential anticancer agent in a few cancer types. However, it is unclear whether and how Til could promote the death of triple-negative breast cancer (TNBC) cells by inducing ferroptosis. Our study determined that Til induced cell death and attenuated cell proliferation in TNBC cells in vitro and in vivo with less toxicity for the first time. Functional assays showed that ferroptosis was the predominant form that contributed to Til-induced cell death of TNBC. Mechanistically, Til induces ferroptosis of TNBC cells via independent PUFA-PLS pathways but is closely involved in the Nrf2/HO-1 pathway. Silencing of HO-1 substantially abrogated the tumor-inhibiting effects of Til. In conclusion, our findings suggest that the natural product Til exerted its antitumor activity on TNBC by promoting ferroptosis, and the HO-1/SLC7A11 pathway plays an indispensable role in Til-induced ferroptotic cell death.
Collapse
Affiliation(s)
- Chuang Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jian-Fu Zhao
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Yi-Ming Wang
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Xian-Lin Wu
- Cancer center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518000, China.
| | - Ling Ye
- Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China.
| |
Collapse
|
38
|
Cheng Z, Chu H, Zhu Q, Yang L. Ferroptosis in non-alcoholic liver disease: Molecular mechanisms and therapeutic implications. Front Nutr 2023; 10:1090338. [PMID: 36992907 PMCID: PMC10040549 DOI: 10.3389/fnut.2023.1090338] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
Ferroptosis refers to a novel modality of regulated cell death characterized by excessive iron accumulation and overwhelming lipid peroxidation, which takes an important part in multiple pathological processes associated with cell death. Considering the crucial roles of the liver in iron and lipid metabolism and its predisposition to oxidative insults, more and more studies have been conducted to explore the relationship between ferroptosis and various liver disorders, including non-alcoholic fatty liver disease (NAFLD). With increased morbidity and high mortality rates, NAFLD has currently emerged as a global public health issue. However, the etiology of NAFLD is not fully understood. In recent years, an accumulating body of evidence have suggested that ferroptosis plays a pivotal role in the pathogenesis of NAFLD, but the precise mechanisms underlying how ferroptosis affects NAFLD still remain obscure. Here, we summarize the molecular mechanisms of ferroptosis and its complicated regulation systems, delineate the different effects that ferroptosis exerts in different stages of NAFLD, and discuss some potential effective therapies targeting ferroptosis for NAFLD treatment, which putatively points out a novel direction for NAFLD treatment.
Collapse
Affiliation(s)
- Zilu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qingjing Zhu
- Jinyintan Hospital, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Qingjing Zhu,
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Ling Yang, ,
| |
Collapse
|
39
|
Ji J, Wu L, Wei J, Wu J, Guo C. The Gut Microbiome and Ferroptosis in MAFLD. J Clin Transl Hepatol 2023; 11:174-187. [PMID: 36406312 PMCID: PMC9647110 DOI: 10.14218/jcth.2022.00136] [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: 03/21/2022] [Revised: 05/22/2022] [Accepted: 06/12/2022] [Indexed: 12/04/2022] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a new disease definition, and is proposed to replace the previous name, nonalcoholic fatty liver disease (NAFLD). Globally, MAFLD/NAFLD is the most common liver disease, with an incidence rate ranging from 6% to 35% in adult populations. The pathogenesis of MAFLD/NAFLD is closely related to insulin resistance (IR), and the genetic susceptibility to acquired metabolic stress-associated liver injury. Similarly, the gut microbiota in MAFLD/NAFLD is being revaluated by scientists, as the gut and liver influence each other via the gut-liver axis. Ferroptosis is a novel form of programmed cell death caused by iron-dependent lipid peroxidation. Emerging evidence suggests that ferroptosis has a key role in the pathological progression of MAFLD/NAFLD, and inhibition of ferroptosis may become a novel therapeutic strategy for the treatment of NAFLD. This review focuses on the main mechanisms behind the promotion of MAFLD/NAFLD occurrence and development by the intestinal microbiota and ferroptosis. It outlines new strategies to target the intestinal microbiota and ferroptosis to facilitate future MAFLD/NAFLD therapies.
Collapse
Affiliation(s)
- Jie Ji
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liwei Wu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jue Wei
- Department of Gastroenterology Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Correspondence to: Chuanyong Guo, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, NO. 301, Middle Yanchang Road, Jing’an District, Shanghai 200072, China. ORCID: https://orcid.org/0000-0002-6527-4673. E-mail: ; Jianye Wu: Department of Gastroenterology, Putuo People’s Hospital, NO. 1291, Jiangning road, Putuo, Shanghai 200060, China. ORCID: https://orcid.org/0000-0003-2675-4241. E-mail:
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Correspondence to: Chuanyong Guo, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, NO. 301, Middle Yanchang Road, Jing’an District, Shanghai 200072, China. ORCID: https://orcid.org/0000-0002-6527-4673. E-mail: ; Jianye Wu: Department of Gastroenterology, Putuo People’s Hospital, NO. 1291, Jiangning road, Putuo, Shanghai 200060, China. ORCID: https://orcid.org/0000-0003-2675-4241. E-mail:
| |
Collapse
|
40
|
Kim K, Yoon H. Gamma-Aminobutyric Acid Signaling in Damage Response, Metabolism, and Disease. Int J Mol Sci 2023; 24:ijms24054584. [PMID: 36902014 PMCID: PMC10003236 DOI: 10.3390/ijms24054584] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Gamma-aminobutyric acid (GABA) plays a crucial role in signal transduction and can function as a neurotransmitter. Although many studies have been conducted on GABA in brain biology, the cellular function and physiological relevance of GABA in other metabolic organs remain unclear. Here, we will discuss recent advances in understanding GABA metabolism with a focus on its biosynthesis and cellular functions in other organs. The mechanisms of GABA in liver biology and disease have revealed new ways to link the biosynthesis of GABA to its cellular function. By reviewing what is known about the distinct effects of GABA and GABA-mediated metabolites in physiological pathways, we provide a framework for understanding newly identified targets regulating the damage response, with implications for ameliorating metabolic diseases. With this review, we suggest that further research is necessary to develop GABA's beneficial and toxic effects on metabolic disease progression.
Collapse
|
41
|
Traber MG, Cross C. Alpha-Tocopherol from people to plants is an essential cog in the metabolic machinery. Antioxid Redox Signal 2023; 38:775-791. [PMID: 36793193 DOI: 10.1089/ars.2022.0212] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
SIGNIFICANCE Protection from oxygen, a di-radical, became a necessity with the evolution of photosynthetic organisms about 2.7 billion years. α-Tocopherol plays an essential role in organisms from plants to people. An overview of human conditions that result in severe vitamin E (α-tocopherol) deficiency is provided. RECENT ADVANCES α-Tocopherol has a critical role in the oxygen protection system by stopping lipid peroxidation, its induced damage and cellular death by ferroptosis. Recent findings in bacteria and plants support the concept of why lipid peroxidation is so dangerous to life and why the family of tocochromanols are essential for aerobic organisms and for plants. CRITICAL ISSUES The hypothesis that prevention of the propagation of lipid peroxidation is the basis for the α-tocopherol requirement in vertebrates is proposed and further that its absence dysregulates energy metabolism, one-carbon metabolism and thiol homeostasis. By recruiting intermediate metabolites from adjacent pathways to sustain effective lipid hydroperoxide elimination, α-tocopherol function is linked not only to NADPH metabolism and its formation through the pentose phosphate pathway via glucose metabolism, but also to sulfur-containing amino acid metabolism, and to one-carbon metabolism. FUTURE DIRECTIONS Evidence from humans, animals and plants support the hypothesis but future studies are needed to assess the genetic sensors that detect lipid peroxidation and cause the ensuing metabolic dysregulation.
Collapse
Affiliation(s)
- Maret G Traber
- Oregon State University, 2694, Linus Pauling Institute, 307 LPSC, Corvallis, Oregon, United States, 97331-4501;
| | - Carroll Cross
- University of California Davis School of Medicine, 12218, Sacramento, California, United States;
| |
Collapse
|
42
|
Zhang J, Sun W, Yan W, Kong X, Shen T, Laubach K, Chen M, Chen X. TP73 Isoform-specific disruption reveals a critical role of TAp73beta in growth suppression and inflammatory response. Cell Death Dis 2023; 14:14. [PMID: 36631448 PMCID: PMC9834251 DOI: 10.1038/s41419-022-05529-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
TP73 is expressed as multiple N- and C-terminal isoforms through two separate promoters or alternative splicing. While N-terminal p73 isoforms have been well studied, very little is known about p73 C-terminal isoforms. Thus, CRISPR was used to delete TP73 Exon13 (E13-KO) to induce p73α to p73β isoform switch. We showed that E13-KO led to decreased cell proliferation and migration and sensitized cells to ferroptosis, which can be reverted by knockdown of TAp73β in E13-KO cells. To understand the biological function of p73β in vivo, we generated a mouse model in that the Trp73 E13 was deleted by CRISPR. We showed that p73α to p73β isoform switch led to increased cellular senescence in mouse embryonic fibroblasts. We also showed that E13-deficient mice exhibited shorter life span and were prone to spontaneous tumors, chronic inflammation and liver steatosis as compared to WT mice. Additionally, we found that the incidence of chronic inflammation and liver steatosis was higher in E13-deficient mice than that in Trp73-deficient mice, suggesting that p73β is a strong inducer of inflammatory response. Mechanistically, we showed that TAp73β was able to induce cysteine dioxygenase 1 (CDO-1), leading to cysteine depletion and subsequently, enhanced ferroptosis and growth suppression. Conversely, knockdown of CDO-1 was able to alleviate the growth suppression and ferroptosis in E13-KO cells. Together, our data suggest that at a physiologically relevant level, TAp73β is a strong inducer of growth suppression but insufficient to compensate for loss of TAp73α in tumor suppression due to aberrant induction of inflammatory response and liver steatosis.
Collapse
Affiliation(s)
- Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC Davis, California, Davis, USA.
| | - Wenqiang Sun
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC Davis, California, Davis, USA
- Department of Animal Science and Technology, Sichuan Agricultural University, Ya'an, China
| | - Wensheng Yan
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC Davis, California, Davis, USA
- Berkeley Regional Lab, Pathology/Lab-Histology Department, The Permanente Medical group, Berkeley, CA, 94085, USA
| | - Xiangmudong Kong
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC Davis, California, Davis, USA
| | - Tong Shen
- West Coast Metabolomics Center, UC Davis, Califronia, Davis, USA
| | - Kyra Laubach
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC Davis, California, Davis, USA
| | - Mingyi Chen
- Department of Pathology, Southwestern Medical Center, University of Texas, Dallas, USA
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC Davis, California, Davis, USA.
| |
Collapse
|
43
|
Treatment of Drug-Induced Liver Injury. Biomedicines 2022; 11:biomedicines11010015. [PMID: 36672522 PMCID: PMC9855719 DOI: 10.3390/biomedicines11010015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/28/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Current pharmacotherapy options of drug-induced liver injury (DILI) remain under discussion and are now evaluated in this analysis. Needless to say, the use of the offending drug must be stopped as soon as DILI is suspected. Normal dosed drugs may cause idiosyncratic DILI, and drugs taken in overdose commonly lead to intrinsic DILI. Empirically used but not substantiated regarding efficiency by randomized controlled trials (RCTs) is the intravenous antidote treatment with N-acetylcysteine (NAC) in patients with intrinsic DILI by N-acetyl-p-aminophenol (APAP) overdose. Good data recommending pharmacotherapy in idiosyncratic DILI caused by hundreds of different drugs are lacking. Indeed, a recent analysis revealed that just eight RCTs have been published, and in only two out of eight trials were DILI cases evaluated for causality by the worldwide used Roussel Uclaf Causality Assessment Method (RUCAM), representing overall a significant methodology flaw, as results of DILI RCTs lacking RUCAM are misleading since many DILI cases are known to be attributable erroneously to nondrug alternative causes. In line with these major shortcomings and mostly based on anecdotal reports, glucocorticoids (GCs) and other immuno-suppressants may be given empirically in carefully selected patients with idiosyncratic DILI exhibiting autoimmune features or caused by immune checkpoint inhibitors (ICIs), while some patients with cholestatic DILI may benefit from ursodeoxycholic acid use; in other patients with drug-induced hepatic sinusoidal obstruction syndrome (HSOS) and coagulopathy risks, the indication for anticoagulants should be considered. In view of many other mechanistic factors such as the hepatic microsomal cytochrome P450 with a generation of reactive oxygen species (ROS), ferroptosis with toxicity of intracellular iron, and modification of the gut microbiome, additional therapy options may be available in the future. In summation, stopping the offending drug is still the first line of therapy for most instances of acute DILI, while various therapies are applied empirically and not based on good data from RCTs awaiting further trials using the updated RUCAM that asks for strict exclusion and inclusion details like liver injury criteria and provides valid causality rankings of probable and highly probable grades.
Collapse
|
44
|
Chen H. Iron metabolism in non-alcoholic fatty liver disease: A promising therapeutic target. LIVER RESEARCH 2022; 6:203-213. [PMID: 39957910 PMCID: PMC11791839 DOI: 10.1016/j.livres.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/05/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common cause of chronic liver disease worldwide, and is closely associated with the increased risk of the prevalence of obesity and diabetes. NAFLD begins with the presence of >5% excessive lipid accumulation in the liver, and potentially develops into non-alcoholic steatohepatitis, fibrosis, cirrhosis and hepatocellular carcinoma. Therefore, insight into the pathogenesis of NAFLD is of key importance to its effective treatment. Iron is an essential element in the life of all mammalian organisms. However, the free iron deposition is positively associated with histological severity in NAFLD patients due to the production of reactive oxygen species via the Fenton reaction. Recently, several iron metabolism-targeted therapies, such as phlebotomy, iron chelators, nanotherapeutics. and ferroptosis, have shown their potential as a therapeutic option in the treatment of NAFLD and as a clinical strategy to intervene in the progression of NAFLD. Herein, we review the recent overall evidence on iron metabolism and provide the mechanism of hepatic iron overload-induced liver pathologies and the recent advances in iron metabolism-targeted therapeutics in the treatment of NAFLD.
Collapse
Affiliation(s)
- Hanqing Chen
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Wu Q, Chen Z, Ding Y, Tang Y, Cheng Y. Protective effect of traditional Chinese medicine on non-alcoholic fatty liver disease and liver cancer by targeting ferroptosis. Front Nutr 2022; 9:1033129. [PMID: 36330148 PMCID: PMC9623008 DOI: 10.3389/fnut.2022.1033129] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease with high incidence and is closely related to metabolic syndrome. If not controlled, it may eventually become hepatocellular carcinoma (HCC). Ferroptosis, a non-apoptotic form of programmed cell death (PCD), is closely related to NAFLD and HCC, and the mechanisms of action involved are more complex. Some studies have demonstrated that many drugs inhibit ferroptosis and protect liver steatosis or carcinogenesis. The role of Traditional Chinese Medicine (TCM), especially herbs or herbal extracts, has received increasing attention. However, there are relatively few review articles on the regulation of NAFLD by TCM through ferroptosis pathway. Here, we summarize the TCM intervention mechanism and application affecting NAFLD/NAFLD-HCC via regulation of ferroptosis. This article focuses on the relationship between ferroptosis and NAFLD or NAFLD-HCC and the protective effect of TCM on both by targeting ferroptosis. It not only summarizes the mechanism of early prevention and treatment of NAFLD, but also provides reference ideas for the development of TCM for the treatment of metabolic diseases and liver diseases.
Collapse
Affiliation(s)
- Qiongbo Wu
- Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, Haikou, China
- Food Science and Technology Center, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Zihao Chen
- Food Science and Technology Center, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Yi Ding
- Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, Haikou, China
| | - Yunting Tang
- Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, Haikou, China
| | - Yawei Cheng
- Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, Haikou, China
- *Correspondence: Yawei Cheng,
| |
Collapse
|
46
|
Teschke R. Aluminum, Arsenic, Beryllium, Cadmium, Chromium, Cobalt, Copper, Iron, Lead, Mercury, Molybdenum, Nickel, Platinum, Thallium, Titanium, Vanadium, and Zinc: Molecular Aspects in Experimental Liver Injury. Int J Mol Sci 2022; 23:12213. [PMID: 36293069 PMCID: PMC9602583 DOI: 10.3390/ijms232012213] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 09/30/2022] [Accepted: 10/11/2022] [Indexed: 11/25/2022] Open
Abstract
Experimental liver injury with hepatocelluar necrosis and abnormal liver tests is caused by exposure to heavy metals (HMs) like aluminum, arsenic, beryllium, cadmium, chromium, cobalt, copper, iron, lead, mercury, molybdenum, nickel, platinum, thallium, titanium, vanadium, and zinc. As pollutants, HMs disturb the ecosystem, and as these substances are toxic, they may affect the health of humans and animals. HMs are not biodegradable and may be deposited preferentially in the liver. The use of animal models can help identify molecular and mechanistic steps leading to the injury. HMs commonly initiate hepatocellular overproduction of ROS (reactive oxygen species) due to oxidative stress, resulting in covalent binding of radicals to macromolecular proteins or lipids existing in membranes of subcellular organelles. Liver injury is facilitated by iron via the Fenton reaction, providing ROS, and is triggered if protective antioxidant systems are exhausted. Ferroptosis syn pyroptosis was recently introduced as mechanistic concept in explanations of nickel (Ni) liver injury. NiCl2 causes increased iron deposition in the liver, upregulation of cyclooxygenase 2 (COX-2) protein and mRNA expression levels, downregulation of glutathione eroxidase 4 (GPX4), ferritin heavy chain 1 (FTH1), nuclear receptor coactivator 4 (NCOA4) protein, and mRNA expression levels. Nickel may cause hepatic injury through mitochondrial damage and ferroptosis, defined as mechanism of iron-dependent cell death, similar to glutamate-induced excitotoxicity but likely distinct from apoptosis, necrosis, and autophagy. Under discussion were additional mechanistic concepts of hepatocellular uptake and biliary excretion of mercury in exposed animals. For instance, the organic anion transporter 3 (Oat3) and the multidrug resistance-associated protein 2 (Mrp2) were involved in the hepatic handling of mercury. Mercury treatment modified the expression of Mrp2 and Oat3 as assessed by immunoblotting, partially explaining its impaired biliary excretion. Concomitantly, a decrease in Oat3 abundance in the hepatocyte plasma membranes was observed that limits the hepatic uptake of mercury ions. Most importantly and shown for the first time in liver injury caused by HMs, titanium changed the diversity of gut microbiota and modified their metabolic functions, leading to increased generation of lipopolysaccharides (LPS). As endotoxins, LPS may trigger and perpetuate the liver injury at the level of gut-liver. In sum, mechanistic and molecular steps of experimental liver injury due to HM administration are complex, with ROS as the key promotional compound. However, additional concepts such as iron used in the Fenton reaction, ferroptosis, modification of transporter systems, and endotoxins derived from diversity of intestinal bacteria at the gut-liver level merit further consideration.
Collapse
Affiliation(s)
- Rolf Teschke
- Department of Internal Medicine II, Division of Gastroenterology and Hepatology, Klinikum Hanau, Academic Teaching Hospital of the Medical Faculty, Goethe University Frankfurt, 63450 Hanau, Germany
| |
Collapse
|
47
|
Ma C, Han L, Zhu Z, Heng Pang C, Pan G. Mineral metabolism and ferroptosis in non-alcoholic fatty liver diseases. Biochem Pharmacol 2022; 205:115242. [PMID: 36084708 DOI: 10.1016/j.bcp.2022.115242] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease worldwide. Minerals including iron, copper, zinc, and selenium, fulfil an essential role in various biochemical processes. Moreover, the identification of ferroptosis and cuproptosis further underscores the importance of intracellular mineral homeostasis. However, perturbation of minerals has been frequently reported in patients with NAFLD and related diseases. Interestingly, studies have attempted to establish an association between mineral disorders and NAFLD pathological features, including oxidative stress, mitochondrial dysfunction, inflammatory response, and fibrogenesis. In this review, we aim to provide an overview of the current understanding of mineral metabolism (i.e., absorption, utilization, and transport) and mineral interactions in the pathogenesis of NAFLD. More importantly, this review highlights potential therapeutic strategies, challenges, future directions for targeting mineral metabolism in the treatment of NAFLD.
Collapse
Affiliation(s)
- Chenhui Ma
- Department of Chemical and Environmental Engineering, University of Nottingham Ningbo China, Ningbo 315100, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Han
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, UK.
| | - Cheng Heng Pang
- Department of Chemical and Environmental Engineering, University of Nottingham Ningbo China, Ningbo 315100, China.
| | - Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
48
|
Li XZ, Xiong ZC, Zhang SL, Hao QY, Gao M, Wang JF, Gao JW, Liu PM. Potential ferroptosis key genes in calcific aortic valve disease. Front Cardiovasc Med 2022; 9:916841. [PMID: 36003913 PMCID: PMC9395208 DOI: 10.3389/fcvm.2022.916841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is a highly prevalent condition that comprises a disease continuum, ranging from microscopic changes to profound fibro-calcific leaflet remodeling, culminating in aortic stenosis, heart failure, and ultimately premature death. Ferroptosis has been hypothesized to contribute to the pathogenesis of CAVD. We aimed to study the association between ferroptosis genes and CAVD and reveal the potential roles of ferroptosis in CAVD. CAVD-related differentially expressed genes (DEGs) were identified via bioinformatic analysis of Datasets GSE51472 and GSE12644 obtained from Gene Expression Omnibus. A ferroptosis dataset containing 259 genes was obtained from the Ferroptosis Database. We then intersected with CAVD-related DEGs to identify the ferroptosis DEGs. Subsequently, protein–protein interaction networks and functional enrichment analyses were performed for ferroptosis DEGs. Then, we used miRWalk3.0 to predict the target pivotal microRNAs. An in vitro model of CAVD was constructed using human aortic valve interstitial cells. The qRT-PCR and western blotting methods were used to validate the ferroptosis DEGs identified by the microarray data. A total of 21 ferroptosis DEGs in CAVD containing 12 upregulated and nine downregulated genes were identified. The results of the Gene Set Enrichment Analysis (GSEA) and analysis of the KEGG pathway by WebGestalt indicated that the ferroptosis DEGs were enriched in six signaling pathways among which NAFLD (including IL-6, BID, and PRKAA2 genes) and HIF-1 (including IL-6, HIF-1, and HMOX1 genes) signaling pathways were also verified by DAVID and/or Metascape. Finally, the in vitro results showed that the mRNA and protein expression levels of IL-6, HIF-1α, HMOX1, and BID were higher, while the levels of PRKAA2 were lower in the Pi-treated group than those in the control group. However, the addition of ferrostatin-1 (a selective ferroptosis inhibitor) significantly reversed the above changes. Therefore, IL-6, HIF-1α, HMOX1, BID, and PRKAA2 are potential key genes closely associated with ferroptosis in CAVD. Further work is required to explore the underlying ferroptosis-related molecular mechanisms and provide possible therapeutic targets for CAVD.
Collapse
Affiliation(s)
- Xiong-Zhi Li
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhuo-Chao Xiong
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shao-Ling Zhang
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qing-Yun Hao
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ming Gao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jing-Feng Wang
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jing-Wei Gao
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jing-Wei Gao
| | - Pin-Ming Liu
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Pin-Ming Liu
| |
Collapse
|
49
|
Patulin disrupts SLC7A11-cystine-cysteine-GSH antioxidant system and promotes renal cell ferroptosis both in vitro and in vivo. Food Chem Toxicol 2022; 166:113255. [PMID: 35772596 DOI: 10.1016/j.fct.2022.113255] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/29/2022] [Accepted: 06/21/2022] [Indexed: 01/10/2023]
Abstract
Patulin (PAT) is a common food-borne mycotoxin with diverse toxic effects including nephrotoxicity. The induction of oxidative stress is suggested to be a key mechanism contributed to toxicities of PAT. Reduced glutathione (GSH), a sulfhydryl-containing tripeptide, is a key reason for PAT-mediated oxidative stress. Cystine/glutamate antiporter (system xc-)-mediated cystine uptake plays a critical role in maintaining redox balance via promoting GSH biosynthesis. In this study, we addressed if GSH reduction by PAT was associated with inhibition of system xc--mediated GSH biosynthesis. Results showed that PAT significantly decreased activity of SLC7A11, a core subunit of system xc-, through activating AMPK-mediated formation of beclin1-SLC7A11 complex. Furthermore, PAT promoted ferroptosis induced by a known ferroptosis inducer RSL3 in normal renal cells, and exacerbated folic acid-induced nephrotoxicity in a mouse model of acute kidney injury. The findings of the present study provide new insights into PAT-induced kidney toxicity, and implicate that patients with ferroptosis-associated diseases maybe more susceptible to PAT.
Collapse
|
50
|
Ferroptosis as a mechanism of non-ferrous metal toxicity. Arch Toxicol 2022; 96:2391-2417. [PMID: 35727353 DOI: 10.1007/s00204-022-03317-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
Abstract
Ferroptosis is a recently discovered form of regulated cell death, implicated in multiple pathologies. Given that the toxicity elicited by some metals is linked to alterations in iron metabolism and induction of oxidative stress and lipid peroxidation, ferroptosis might be involved in such toxicity. Although direct evidence is insufficient, certain pioneering studies have demonstrated a crosstalk between metal toxicity and ferroptosis. Specifically, the mechanisms underlying metal-induced ferroptosis include induction of ferritinophagy, increased DMT-1 and TfR cellular iron uptake, mitochondrial dysfunction and mitochondrial reactive oxygen species (mitoROS) generation, inhibition of Xc-system and glutathione peroxidase 4 (GPX4) activity, altogether resulting in oxidative stress and lipid peroxidation. In addition, there is direct evidence of the role of ferroptosis in the toxicity of arsenic, cadmium, zinc, manganese, copper, and aluminum exposure. In contrast, findings on the impact of cobalt and nickel on ferroptosis are scant and nearly lacking altogether for mercury and especially lead. Other gaps in the field include limited studies on the role of metal speciation in ferroptosis and the critical cellular targets. Although further detailed studies are required, it seems reasonable to propose even at this early stage that ferroptosis may play a significant role in metal toxicity, and its modulation may be considered as a potential therapeutic tool for the amelioration of metal toxicity.
Collapse
|