201
|
Hua Y, Yang S, Zhang Y, Li J, Wang M, Yeerkenbieke P, Liao Q, Liu Q. Modulating ferroptosis sensitivity: environmental and cellular targets within the tumor microenvironment. J Exp Clin Cancer Res 2024; 43:19. [PMID: 38217037 PMCID: PMC10787430 DOI: 10.1186/s13046-023-02925-5] [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: 09/11/2023] [Accepted: 12/06/2023] [Indexed: 01/14/2024] Open
Abstract
Ferroptosis, a novel form of cell death triggered by iron-dependent phospholipid peroxidation, presents significant therapeutic potential across diverse cancer types. Central to cellular metabolism, the metabolic pathways associated with ferroptosis are discernible in both cancerous and immune cells. This review begins by delving into the intricate reciprocal regulation of ferroptosis between cancer and immune cells. It subsequently details how factors within the tumor microenvironment (TME) such as nutrient scarcity, hypoxia, and cellular density modulate ferroptosis sensitivity. We conclude by offering a comprehensive examination of distinct immunophenotypes and environmental and metabolic targets geared towards enhancing ferroptosis responsiveness within the TME. In sum, tailoring precise ferroptosis interventions and combination strategies to suit the unique TME of specific cancers may herald improved patient outcomes.
Collapse
Affiliation(s)
- Yuze Hua
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Sen Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yalu Zhang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
- Department of General Surgery, Anhui Provincial Hospital, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230027, China
| | - Jiayi Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Mengyi Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Palashate Yeerkenbieke
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
- Department of General Surgery, Xinjiang Yili Kazak Autonomous Prefecture Friendship Hospital, Xinjiang, 835099, China
| | - Quan Liao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| | - Qiaofei Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
202
|
Hu Y, Li Q, Wang Y. Serum ACSL4 levels in patients with ST-segment elevation myocardial infarction (STEMI) and its association with one-year major adverse cardiovascular events (MACE): A prospective cohort study. Medicine (Baltimore) 2024; 103:e36870. [PMID: 38215103 PMCID: PMC10783377 DOI: 10.1097/md.0000000000036870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/15/2023] [Indexed: 01/14/2024] Open
Abstract
In the present prospective cohort research, we aimed to explore the serum levels of Acyl-CoA synthetase long-chain family member 4 (ACSL4) in patients with ST-segment elevation myocardial infarction (STEMI) and its association with 1-year major adverse cardiovascular events (MACE). This prospective cohort study recruited 507 patients who underwent percutaneous coronary intervention for the treatment of STEMI at our hospital during August 2019 to July 2022. The serum ACSL4, tumor necrosis factor-α, interleukin (IL)-6, IL-1β, and C-reactive protein levels were measured by enzyme-linked immunosorbent assay. Demographic and clinical statistics were also collected. In addition, all patients were followed up for 1 year, and patients with MACE were defined as poor prognosis group. All data used SPSS 26.0 to statistical analyses. The poor prognosis group had significantly higher age and low-density leptin cholesterol (LDLC) levels compared to the favorable prognosis group (P < .05). STEMI patients exhibited significantly elevated serum levels of ACSL4, tumor necrosis factor-α, IL-6, IL-1β, and C-reactive protein (P < .05). Serum ACSL4 and IL-1β levels in the poor prognosis group were remarkably enhanced compared to the favorable prognosis group. Curvilinear regression analysis demonstrated that ACSL4 was associated with LDLC and IL-1β. Moreover, ACSL4 (B = 0.138, 95% CI 1.108-1.189, P < .001), LDLC (B = 2.317, 95% CI 5.253-19.603, P < .001), and IL-1β (B = 0.061, 95%CI 1.008-1.122, P = .025) levels were the risk factors for STEMI patients with 1-year MACE. This study showed that the serum ACSL4 levels was remarkably elevated in STEMI patients. This study might provide new targets and a comprehensive approach to cardiovascular protection in STEMI patients.
Collapse
Affiliation(s)
- Yun Hu
- Emergency Department, Wuhan Dongxihu District People’s Hospital, Wuhan, China
| | - Qingye Li
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yinglin Wang
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
203
|
Zhou D, Lu P, Mo X, Yang B, Chen T, Yao Y, Xiong T, Yue L, Yang X. Ferroptosis and metabolic syndrome and complications: association, mechanism, and translational applications. Front Endocrinol (Lausanne) 2024; 14:1248934. [PMID: 38260171 PMCID: PMC10800994 DOI: 10.3389/fendo.2023.1248934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Metabolic syndrome is a medical condition characterized by several metabolic disorders in the body. Long-term metabolic disorders raise the risk of cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM). Therefore, it is essential to actively explore the aetiology of metabolic syndrome (MetS) and its comorbidities to provide effective treatment options. Ferroptosis is a new form of cell death that is characterized by iron overload, lipid peroxide accumulation, and decreased glutathione peroxidase 4(GPX4) activity, and it involves the pathological processes of a variety of diseases. Lipid deposition caused by lipid diseases and iron overload is significant in metabolic syndrome, providing the theoretical conditions for developing ferroptosis. Recent studies have found that the major molecules of ferroptosis are linked to common metabolic syndrome consequences, such as T2DM and atherosclerosis. In this review, we first discussed the mechanics of ferroptosis, the regulatory function of inducers and inhibitors of ferroptosis, and the significance of iron loading in MetS. Next, we summarized the role of ferroptosis in the pathogenesis of MetS, such as obesity, type 2 diabetes, and atherosclerosis. Finally, we discussed relevant ferroptosis-targeted therapies and raised some crucial issues of concern to provide directions for future Mets-related treatments and research.
Collapse
Affiliation(s)
- Dongmei Zhou
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Peipei Lu
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xianglai Mo
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Bing Yang
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Ting Chen
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - You Yao
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Tian Xiong
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Lin Yue
- School of Nursing, Hunan University of Medicine, Huaihua, China
| | - Xi Yang
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| |
Collapse
|
204
|
Wang S, Li X, Li J, Wang A, Li F, Hu H, Long T, Pei X, Li H, Zhong F, Zhu F. Inhibition of cisplatin-induced Acsl4-mediated ferroptosis alleviated ovarian injury. Chem Biol Interact 2024; 387:110825. [PMID: 38056807 DOI: 10.1016/j.cbi.2023.110825] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/05/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Given that the severity of the chemotherapy-induced ovarian damage, effective fertility preservation is a necessary part of the treatment process. Ferroptosis is a regulated cell death triggered by excessive phospholipid peroxidation caused by iron and the role of ferroptosis in chemotherapy-induced ovarian damage remains unclear. In this study, we demonstrated that cisplatin treatment caused the accumulation of iron ions which induced ferroptosis in ovarian tissue. And our results show that ferrostatin-1 was able to suppress the ovarian injury and granulosa cell death caused by cisplatin (Cis) in vivo and in vitro. At the same time, we observed significant changes in the expression levels of Acyl-CoA synthetase long-chain family member 4 (Acsl4) and glutathione peroxidase 4 (GPX4). Similarly, Rosiglitazone, an inhibitor of Acsl4, administration alleviated the ovary damage of the mice undergoing chemotherapy. Further mechanistic investigation showed that cisplatin increased the expression level of specificity protein 1 (SP1), and SP1 could bind to the promoter of Acsl4 to increased Acsl4 transcription. Overall, ferroptosis plays an important role in Cis induced ovarian injury, and inhibition of ferroptosis protects ovarian tissues from damage caused by cisplatin, and for the first time, we have identified the potential of Fer-1 and Rosi to protect ovarian function in female mice undergoing chemotherapy.
Collapse
Affiliation(s)
- Siyuan Wang
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Xuqing Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Jun Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Aiping Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Fangfang Li
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Huiqing Hu
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Tengfei Long
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Xueting Pei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Hongyan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China.
| | - Fei Zhong
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China.
| | - Fengyu Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
205
|
Xiong T, Wang Y, Zhu C. A risk model based on 10 ferroptosis regulators and markers established by LASSO-regularized linear Cox regression has a good prognostic value for ovarian cancer patients. Diagn Pathol 2024; 19:4. [PMID: 38178187 PMCID: PMC10765690 DOI: 10.1186/s13000-023-01414-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/12/2023] [Indexed: 01/06/2024] Open
Abstract
Ovarian cancer is the deadliest gynecologic cancer due to its high rate of recurrence and limited early diagnosis. For certain patients, particularly those with recurring disorders, standard treatment alone is insufficient in the majority of cases. Ferroptosis, an iron- and ROS (reactive oxygen species)-reliant cell death, plays a vital role in the occurrence of ovarian cancer. Herein, subjects from TCGA-OV were calculated for immune scores using the ESTIMATE algorithm and assigned into high- (N = 185) or low-immune (N = 193) score groups; 259 ferroptosis regulators and markers were analyzed for expression, and 64 were significantly differentially expressed between two groups. These 64 differentially expressed genes were applied for LASSO-regularized linear Cox regression for establishing ferroptosis regulators and a markers-based risk model, and a 10-gene signature was established. The ROC curve indicated that the risk score-based curve showed satisfactory predictive efficiency. Univariate and multivariate Cox risk regression analyses showed that age and risk score were risk factors for ovarian cancer patients' overall survival; patients in the high-risk score group obtained lower immune scores. The Nomogram analysis indicated that the model has a good prognostic performance. GO functional enrichment annotation confirmed again the involvement of these 10 genes in ferroptosis and immune activities. TIMER online analysis showed that risk factors and immune cells were significantly correlated. In conclusion, the risk model based on 10 ferroptosis regulators and markers has a good prognostic value for ovarian cancer patients.
Collapse
Affiliation(s)
- Tingchuan Xiong
- Department of Gynecologic Surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, 830011, China
| | - Yinghong Wang
- Center of Heath Management, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Changjun Zhu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, 300387, China.
- Laboratory of Molecular and Cellular Systems Biology, College of Life Science, Tianjin Normal University, Tianjin, 300387, China.
| |
Collapse
|
206
|
Feng J, Chen H, Liu Y, Ai Q, Yang Y, He W, Zhao L, Chu S, Chen N. Ferroptosis is Involved in the Pharmacological Effect of Ginsenoside. Mini Rev Med Chem 2024; 24:1228-1237. [PMID: 38213172 PMCID: PMC11337240 DOI: 10.2174/0113895575277359231210145922] [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: 09/02/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 01/13/2024]
Abstract
Ginsenoside is the principal active ingredient in ginseng. Several investigations have found that ginsenosides have anti-inflammatory, antioxidant, anti-apoptotic, anti-cancer, and antiallergic activities. Ferroptosis is an iron-dependent, non-apoptotic form of cell-regulated death caused by lipid peroxidation. Iron, lipid, and amino acid metabolism orchestrate the complex ferroptosis response through direct or indirect regulation of iron accumulation or lipid peroxidation. More and more research has demonstrated that ginsenoside impacts illnesses via ferroptosis, implying that ferroptosis might be employed as a novel target of ginsenoside for disease therapy. This article examines the molecular mechanism of ferroptosis as well as the current advancement of ginsenoside in influencing disorders via ferroptosis.
Collapse
Affiliation(s)
- Juling Feng
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China
- Research lab of translational medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Haodong Chen
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China
| | - Yangbo Liu
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China
| | - Qidi Ai
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China
| | - Yantao Yang
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China
| | - Wenbin He
- Shanxi Key Laboratory of Traditional Chinese Medicine Encephalopathy, Shanxi University of Traditional Chinese Medicine, Taiyuan, 030024, China
| | - Lei Zhao
- Research lab of translational medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Naihong Chen
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
207
|
Inagaki NF, Nakanishi H, Ohto T, Shindou H, Shimizu T. LPCAT3/LPLAT12 deficiency in the liver ameliorates acetaminophen-induced acute liver injury. FASEB J 2024; 38:e23328. [PMID: 38019192 DOI: 10.1096/fj.202301744r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023]
Abstract
Acetaminophen (APAP) is a double-edged sword, mainly depending on the dosage. A moderate dose of APAP is effective for fever and pain relief; however, an overdose induces acute liver injury. The mechanism underlying APAP-induced acute liver failure is unclear, and its treatment is limited. A recent report has shown that several oxidized phospholipids are associated with APAP-induced acute liver failure. Lysophosphatidylcholine acyltransferase 3 (Lpcat3, Lplat12), which is highly expressed in the liver, preferentially catalyzes the incorporation of arachidonate into lysophospholipids (PLs). In the present study, we investigated the roles of Lpcat3 on APAP-induced acute liver injury using liver-specific Lpcat3-knockout mice. Hepatic Lpcat3 deficiency reduced the degree of APAP-induced necrosis of hepatocytes around Zone 3 and ameliorated the elevation of hepatic injury serum marker levels, and prolonged survival. Lipidomic analysis showed that the accumulation of oxidized and hydroperoxidized phospholipids was suppressed in Lpcat3-knockout mice. The amelioration of APAP-induced acute liver injury was due not only to the reduction in the lipid synthesis of arachidonic acid PLs because of Lpcat3 deficiency, but also to the promotion of the APAP detoxification pathway by facilitating the conjugation of glutathione and N-acetyl-p-benzoquinone imine. Our findings suggest that Lpcat3 is a potential therapeutic target for treating APAP-induced acute liver injury.
Collapse
Affiliation(s)
- Natsuko F Inagaki
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
| | | | | | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Lipid Life Science, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
- Institute of Microbial Chemistry, Tokyo, Japan
| |
Collapse
|
208
|
Li S, Kang Y, Zeng Y. Targeting tumor and bone microenvironment: Novel therapeutic opportunities for castration-resistant prostate cancer patients with bone metastasis. Biochim Biophys Acta Rev Cancer 2024; 1879:189033. [PMID: 38040267 DOI: 10.1016/j.bbcan.2023.189033] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/22/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
Despite standard hormonal therapy that targets the androgen receptor (AR) attenuates prostate cancer (PCa) effectively in the initial stage, the tumor ultimately converts to castration-resistant prostate cancer (CRPC), and the acquired resistance is still a great challenge for the management of advanced prostate cancer patients. The tumor microenvironment (TME) consists of multiple cellular and noncellular agents is well known as a vital role during the development and progression of CRPC by establishing communication between TME and tumor cells. Additionally, as primary prostate cancer progresses towards metastasis, and CRPC always experiences bone metastasis, the TME is conducive to the spread of tumors to the distant sits, particularly in bone. In addition, the bone microenvironment (BME) is also closely related to the survival, growth and colonization of metastatic tumor cells. The present review summarized the recent studies which mainly focused on the role of TME or BME in the CRPC patients with bone metastasis, and discussed the underlying mechanisms, as well as the potential therapeutic values of targeting TME and BME in the management of metastatic CRPC patients.
Collapse
Affiliation(s)
- Shenglong Li
- Second ward of Bone and Soft Tissue Tumor Surgery,Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China; The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, Shenyang, China
| | - Yue Kang
- Department of Breast Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yu Zeng
- Department of Urology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China.
| |
Collapse
|
209
|
Chu C, Wang X, Chen F, Yang C, Shi L, Xu W, Wang K, Liu B, Wang C, Sun D, Li J, Ding W. Neutrophil extracellular traps aggravate intestinal epithelial necroptosis in ischaemia-reperfusion by regulating TLR4/RIPK3/FUNDC1-required mitophagy. Cell Prolif 2024; 57:e13538. [PMID: 37691112 PMCID: PMC10771116 DOI: 10.1111/cpr.13538] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Neutrophil extracellular trap (NET) has been confirmed to be related to gut barrier injury during intestinal ischaemia-reperfusion (II/R). However, the specific molecular regulatory mechanism of NETs in II/R-induced intestinal barrier damage has yet to be fully elucidated. Here, we reported increased NETs infiltration accompanied by elevated inflammatory cytokines, cellular necroptosis and tight junction disruption in the intestine of human II/R patients. Meanwhile, NETs aggravated Caco-2 intestinal epithelial cell necroptosis, impairing the monolayer barrier in vitro. Moreover, Pad4-deficient mice were used further to validate the role of NETs in II/R-induced intestinal injury. In contrast, NET inhibition via Pad4 deficiency alleviated intestinal inflammation, attenuated cellular necroptosis, improved intestinal permeability, and enhanced tight junction protein expression. Notably, NETs prevented FUN14 domain-containing 1 (FUNDC1)-required mitophagy activation in intestinal epithelial cells, and stimulating mitophagy attenuated NET-associated mitochondrial dysfunction, cellular necroptosis, and intestinal damage. Mechanistically, silencing Toll-like receptor 4 (TLR4) or receptor-interacting protein kinase 3 (RIPK3) via shRNA relieved mitophagy limitation, restored mitochondrial function and reduced NET-induced necroptosis in Caco-2 cells, whereas this protective effect was reversed by TLR4 or RIPK3 overexpression. The regulation of TLR4/RIPK3/FUNDC1-required mitophagy by NETs can potentially induce intestinal epithelium necroptosis.
Collapse
Affiliation(s)
- Chengnan Chu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Xinyu Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Fang Chen
- Division of Trauma and Acute Care Surgery, Jinling Hospital, School of MedicineSoutheast UniversityNanjingJiangsu ProvinceChina
| | - Chao Yang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Lin Shi
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical EngineeringNanjing University of Science and TechnologyNanjingJiangsu ProvinceChina
| | - Weiqi Xu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Kai Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Baochen Liu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Chenyang Wang
- Key Laboratory of Intestinal Injury, Research Institute of General Surgery, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingJiangsuChina
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical EngineeringNanjing University of Science and TechnologyNanjingJiangsu ProvinceChina
| | - Jieshou Li
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Weiwei Ding
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
- Division of Trauma and Acute Care Surgery, Jinling Hospital, School of MedicineSoutheast UniversityNanjingJiangsu ProvinceChina
| |
Collapse
|
210
|
Chen Y, Zhao J, Ye H, Ceylan-Isik AF, Zhang B, Liu Q, Yang Y, Dong M, Luo B, Ren J. Beneficial impact of cardiac heavy metal scavenger metallothionein in sepsis-provoked cardiac anomalies dependent upon regulation of endoplasmic reticulum stress and ferroptosis but not autophagy. Life Sci 2024; 336:122291. [PMID: 38030060 DOI: 10.1016/j.lfs.2023.122291] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/21/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
AIMS Sepsis represents a profound proinflammatory response with a major contribution from oxidative injury. Here we evaluated possible impact of heavy metal scavenger metallothionein (MT) on endotoxin lipopolysaccharide (LPS)-induced oxidative stress, endoplasmic reticulum (ER) stress, autophagy, and ferroptosis enroute to myocardial injury along with interplay among these stress domains. MATERIALS AND METHODS Echocardiographic, cardiomyocyte mechanical and intracellular Ca2+ responses were monitored in myocardia from WT and transgenic mice with cardiac-selective MT overexpression challenged with LPS. Oxidative stress, stress signaling (p38, ERK, JNK), ER stress, autophagy, and ferroptosis were scrutinized. KEY FINDINGS RNAseq analysis revealed discrepant patterns in ferroptosis between LPS-exposed and normal murine hearts. LPS insult enlarged LV end systolic dimension, suppressed fractional shortening, ejection fraction, maximal velocity of shortening/relengthening and peak shortening, as well as elongated relengthening along with dampened intracellular Ca2+ release and reuptake. In addition, LPS triggered oxidative stress (lowered glutathione/glutathione disulfide ratio and O2- production), activation of stress cascades (p38, ERK, JNK), ER stress (GRP78, PERK, Gadd153, and IRE1α), inflammation (TNFα and iNOS), unchecked autophagy (LCB3, Beclin-1 and Atg7), ferroptosis (GPx4 and SLC7A11) and interstitial fibrosis. Although MT overexpression itself did not reveal response on cardiac function, it attenuated or mitigated LPS-evoked alterations in echocardiographic, cardiomyocyte contractile and intracellular Ca2+ characteristics, O2- production, TNFα level, ER stress and ferroptosis (without affecting autophagy, elevated AMP/ATP ratio, and iNOS). In vitro evidence revealed beneficial effects of suppression of oxidative stress, ER stress and ferroptosis against LPS-elicited myocardial anomalies. SIGNIFICANCE These data strongly support the therapeutic promises of MT and ferroptosis in septic cardiomyopathy.
Collapse
Affiliation(s)
- Yuanzhuo Chen
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Jian Zhao
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Hua Ye
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Burns & Plastic and Wound Repair, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Asli F Ceylan-Isik
- Ankara Yildirim Beyazit University, Faculty of Medicine, Department of Medical Pharmacology, Bilkent, Ankara, Turkey
| | - Bingfang Zhang
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Qiong Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi 710069, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi 710069, China
| | - Maolong Dong
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bijun Luo
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
211
|
Xu Y, Li K, Zhao Y, Zhou L, He N, Qiao H, Xu Q, Zhang H, Liu Y, Zhao J. Inhibition of 15-hydroxyprostaglandin dehydrogenase protects neurons from ferroptosis in ischemic stroke. MedComm (Beijing) 2024; 5:e452. [PMID: 38188604 PMCID: PMC10771813 DOI: 10.1002/mco2.452] [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: 03/06/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 01/09/2024] Open
Abstract
Ischemic stroke is an acute serious cerebrovascular disease with high mortality and disability. Ferroptosis is an important regulated cell death (RCD) in ischemic stroke. 15-Hydroxyprostaglandin dehydrogenase (15-PGDH), a degrading enzyme of prostaglandin E2 (PGE2), is shown to regulate RCD such as autophagy and apoptosis. The study aimed to determine whether 15-PGDH regulates ferroptosis and ischemic stroke, and further the exact mechanism. We demonstrated that overexpression of 15-PGDH in the brain tissues or primary cultured neurons significantly aggravated cerebral injury and neural ferroptosis in ischemic stroke. While inhibition of 15-PGDH significantly protected against cerebral injury and neural ferroptosis, which benefits arise from the activation of the PGE2/PGE2 receptor 4 (EP4) axis. While the impact of 15-PGDH was abolished with glutathione peroxidase 4 (GPX4) deficiency. Then, 15-PGDH inhibitor was found to promote the activation of cAMP-response element-binding protein (CREB) and nuclear factor kappa-B (NF-κB) via the PGE2/EP4 axis, subsequently transcriptionally upregulate the expression of GPX4. In summary, our study indicates that inhibition of 15-PGDH promotes the activation PGE2/EP4 axis, subsequently transcriptionally upregulates the expression of GPX4 via CREB and NF-κB, and then protects neurons from ferroptosis and alleviates the ischemic stroke. Therefore, 15-PGDH may be a potential therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Yunfei Xu
- Department of PathophysiologySchool of Basic Medical SciencesCentral South UniversityChangshaHunanChina
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
- Postdoctoral Research Station of BiologySchool of Basic Medical ScienceCentral South UniversityChangshaHunanChina
| | - Kexin Li
- Department of PathophysiologySchool of Basic Medical SciencesCentral South UniversityChangshaHunanChina
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
| | - Yao Zhao
- Department of PathophysiologySchool of Basic Medical SciencesCentral South UniversityChangshaHunanChina
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
| | - Lin Zhou
- Department of PathophysiologySchool of Basic Medical SciencesCentral South UniversityChangshaHunanChina
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
| | - Nina He
- Department of PathophysiologySchool of Basic Medical SciencesCentral South UniversityChangshaHunanChina
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
| | - Haoduo Qiao
- Department of PathophysiologySchool of Basic Medical SciencesCentral South UniversityChangshaHunanChina
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
| | - Qing Xu
- Department of PathophysiologySchool of Basic Medical SciencesCentral South UniversityChangshaHunanChina
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
| | - Huali Zhang
- Department of PathophysiologySchool of Basic Medical SciencesCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
| | - Ying Liu
- Department of PathophysiologySchool of Basic Medical SciencesCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
| | - Jie Zhao
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Sepsis Translational Medicine Key Lab of Hunan ProvinceChangshaHunanChina
- National Medicine Functional Experimental Teaching CenterCentral South UniversityChangshaHunanChina
| |
Collapse
|
212
|
Yu Z, Mo Z, Qiu Y, Lu H, Zheng B, Liu L. Emerging Therapeutic Approaches Targeting Ferroptosis in Cancer: Focus on Immunotherapy and Nanotechnology. Curr Pharm Biotechnol 2024; 25:2012-2021. [PMID: 38284738 DOI: 10.2174/0113892010276664231228124157] [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: 09/04/2023] [Revised: 11/25/2023] [Accepted: 12/11/2023] [Indexed: 01/30/2024]
Abstract
Ferroptosis is a newly discovered form of programmed cell death characterized by iron overload, ROS accumulation, and lipid peroxidation. It is distinguished by unique morphological, biochemical, and genetic features and stands apart from other known regulated cell death mechanisms. Studies have demonstrated a close association between ferroptosis and various cancers, including liver cancer, lung cancer, renal cell carcinoma, colorectal cancer, pancreatic cancer, and ovarian cancer. Inducing ferroptosis has shown promising results in inhibiting tumor growth and reversing tumor progression. However, the challenge lies in regulating ferroptosis in vivo due to the scarcity of potent compounds that can activate it. Integrating emerging biomedical discoveries and technological innovations with conventional therapies is imperative. Notably, considerable progress has been made in cancer treatment by leveraging immunotherapy and nanotechnology to trigger ferroptosis. This review explores the relationship between ferroptosis and emerging immunotherapies and nanotechnologies, along with their potential underlying mechanisms, offering valuable insights for developing novel cancer treatment strategies.
Collapse
Affiliation(s)
- Zongchao Yu
- Department of General Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhongcheng Mo
- Department of Histology and Embryology, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Yuan Qiu
- Department of General Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hengzhe Lu
- Department of General Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Biao Zheng
- Department of Histology and Embryology, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Longfei Liu
- Department of General Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
213
|
Lu Y, Shao Y, Cui W, Jia Z, Zhang Q, Zhao Q, Chen Z, Yan J, Chu B, Yuan J. Excessive Lipid Peroxidation in Uterine Epithelium Causes Implantation Failure and Pregnancy Loss. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302887. [PMID: 38044324 PMCID: PMC10811501 DOI: 10.1002/advs.202302887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 09/06/2023] [Indexed: 12/05/2023]
Abstract
The uterine epithelium undergoes a dramatic spatiotemporal transformation to enter a receptive state, involving a complex interaction between ovarian hormones and signals from stromal and epithelial cells. Redox homeostasis is critical for cellular physiological steady state; emerging evidence reveals that excessive lipid peroxides derail redox homeostasis, causing various diseases. However, the role of redox homeostasis in early pregnancy remains largely unknown. It is found that uterine deletion of Glutathione peroxidase 4 (GPX4), a key factor in repairing oxidative damage to lipids, confers defective implantation, leading to infertility. To further pinpoint Gpx4's role in different cell types, uterine epithelial-specific Gpx4 is deleted by a lactotransferrin (Ltf)-Cre driver; the resultant females are infertile, suggesting increased lipid peroxidation levels in uterine epithelium compromises receptivity and implantation. Lipid peroxidation inhibitor administration failed to rescue implantation due to carbonylation of major receptive-related proteins underlying high lipid reactive oxygen species. Intriguingly, superimposition of Acyl-CoA synthetase long-chain family member 4 (ACSL4), an enzyme that promotes biosynthesis of phospholipid hydroperoxides, along with uterine epithelial GPX4 deletion, preserves reproductive capacity. This study reveals the pernicious impact of unbalanced redox signaling on embryo implantation and suggests the obliteration of lipid peroxides as a possible therapeutic approach to prevent implantation defects.
Collapse
Affiliation(s)
- Yafang Lu
- Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Yuhan Shao
- Center for Reproductive MedicineShandong UniversityJinanShandong250021China
- Key Laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandong250021China
| | - Weiwei Cui
- Department of Cell BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Zhaoyu Jia
- Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Qian Zhang
- Center for Reproductive MedicineShandong UniversityJinanShandong250021China
- Key Laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandong250021China
| | - Qing Zhao
- Center for Reproductive MedicineShandong UniversityJinanShandong250021China
- Key Laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandong250021China
| | - Zi‐Jiang Chen
- Center for Reproductive MedicineShandong UniversityJinanShandong250021China
- Key Laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandong250021China
| | - Junhao Yan
- Center for Reproductive MedicineShandong UniversityJinanShandong250021China
- Key Laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandong250021China
| | - Bo Chu
- Department of Cell BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Jia Yuan
- Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinanShandong250012China
| |
Collapse
|
214
|
Ma Y, Huang L, Zhang Z, Yang P, Chen Q, Zeng X, Tan F, Wang C, Ruan X, Liao X. CD36 promotes tubular ferroptosis by regulating the ubiquitination of FSP1 in acute kidney injury. Genes Dis 2024; 11:449-463. [PMID: 37588197 PMCID: PMC10425750 DOI: 10.1016/j.gendis.2022.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/20/2022] [Accepted: 12/01/2022] [Indexed: 01/05/2023] Open
Abstract
Reactive oxidative species (ROS) production-driven ferroptosis plays a role in acute kidney injury (AKI). However, its exact molecular mechanism is poorly understood. Scavenger receptor CD36 has important roles in oxidizing lipids, lipid accumulation, metabolic syndrome, and insulin resistance in chronic kidney disease, but its roles remain unexplored in AKI. The present study investigated the role and mechanism of CD36 in regulating proximal tubular cell ferroptosis and AKI. The expression of CD36 was found to be significantly up-regulated in AKI renal tissues and correlated with renal function, which might serve as an independent biomarker for AKI patients. Moreover, in adult mice subjected to AKI, deletion of CD36 (CD36-/-) induced tubular cell ROS accumulation, ferroptosis activation, and renal injury. Mechanistically, combining LC-MS/MS, co-IP, and ubiquitination analyses revealed that CD36 could specifically bind to ferroptosis suppressor protein 1 (FSP1) and regulate its ubiquitination at sites K16 and K24, leading to FSP1 degradation and progression of ferroptosis in AKI. The present results emphasize a novel mechanism of CD36 in cisplatin-induced AKI. The discovery of the special CD36 roles in promoting ferroptosis and AKI development by regulating the ubiquitination of FSP1 in proximal tubular cells may be potential therapeutic targets for AKI. Moreover, CD36 may play a key role in the progression of AKI. Therefore, targeting CD36 may provide a promising treatment option for AKI.
Collapse
Affiliation(s)
- Yixin Ma
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Lili Huang
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Zheng Zhang
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing 400016, China
| | - Pengfei Yang
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Qingsong Chen
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Xujia Zeng
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Fangyan Tan
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Chunxia Wang
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Xiongzhong Ruan
- Centre for Nephrology, Royal Free and University College Medical School, University College London, Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom
- Centre for Lipid Research, Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
- Kuanren Laboratory of Translational Lipidology, Centre for Lipid Research, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiaohui Liao
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
- Kuanren Laboratory of Translational Lipidology, Centre for Lipid Research, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
215
|
Tang D, Kroemer G, Kang R. Ferroptosis in immunostimulation and immunosuppression. Immunol Rev 2024; 321:199-210. [PMID: 37424139 DOI: 10.1111/imr.13235] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/11/2023]
Abstract
Ferroptosis is a form of iron-dependent regulated cell death characterized by the accumulation of toxic lipid peroxides, particularly in the plasma membrane, leading to lytic cell death. While it plays a crucial role in maintaining the overall health and proper functioning of multicellular organisms, it can also contribute to tissue damage and pathological conditions. Although ferroptotic damage is generally recognized as an immunostimulatory process associated with the release of damage-associated molecular patterns (DAMPs), the occurrence of ferroptosis in immune cells or the release of immunosuppressive molecules can result in immune tolerance. Consequently, there is ongoing exploration of targeting the upstream signals or the machinery of ferroptosis to therapeutically enhance or inhibit the immune response. In addition to introducing the core molecular mechanisms of ferroptosis, we will focus on the immune characteristics of ferroptosis in pathological conditions, particularly in the context of infection, sterile inflammation, and tumor immunity.
Collapse
Affiliation(s)
- Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
216
|
Liang Y, Qiu S, Zou Y, Luo L. Targeting ferroptosis with natural products in liver injury: new insights from molecular mechanisms to targeted therapies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155134. [PMID: 37863001 DOI: 10.1016/j.phymed.2023.155134] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Ferroptosis is a brand-new type of controlled cell death that is distinguished by its reliance on iron and the production of lipid peroxidation. The role of ferroptosis in damaging liver disorders has attracted a lot of attention in recent years. One effective strategy to reduce liver damage is to target ferroptosis. PURPOSE The purpose of this review is to clarify the connection between ferroptosis and liver damage and to look into the potential contribution of natural products to the clinical management of liver damage and the discovery of novel medications. METHODS To study the methods by which natural products operate on ferroptosis to cure liver damage and their main signaling pathways, we searched databases from the time of initial publication to August 2023 in PubMed, EMBASE, Web of Science, Ovid, ScienceDirect, and China National Knowledge Infrastructure. The liver illness that each natural product treats is categorized and summarized. It's interesting to note that several natural compounds, such Artemether, Fucoidan sulfate, Curcumin, etc., have the benefit of having many targets and multiple pathways of action. RESULTS We saw that in human samples or animal models of liver injury, ferroptosis indicators were activated, lipid peroxidation levels were elevated, and iron inhibitors had the ability to reduce liver damage. Liver damage can be treated with natural products by regulating ferroptosis. This is mostly accomplished through the modulation of Nrf2-related pathways (e.g., Conclusions and Astaxanthin), biological enzymes like GPX4 and the SIRT family (e.g., Chrysophanol and Decursin), and transcription factors like P53 (e.g., Artemether and Zeaxanthin). CONCLUSIONS This review proposes a promising path for the therapeutic therapy of liver damage by providing a theoretical foundation for the management of ferroptosis utilizing natural ingredients.
Collapse
Affiliation(s)
- Yongyi Liang
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Shaojun Qiu
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Youwen Zou
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, Guangdong, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, 524023, Guangdong, China.
| |
Collapse
|
217
|
Fu Y, Song Y, Jiang D, Pan J, Li W, Zhang X, Chen W, Tian Y, Shen X, Huang Y. Comprehensive Transcriptomic and Metabolomic Analysis Revealed the Functional Differences in Pigeon Lactation between Male and Female during the Reproductive Cycle. Animals (Basel) 2023; 14:75. [PMID: 38200806 PMCID: PMC10778231 DOI: 10.3390/ani14010075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Lactation is a unique reproductive behavior in pigeons, with the crop serving as the organ responsible for secreting pigeon milk. Both male and female pigeons can produce crop milk and rear their offspring through a division of labor. Since the time of the secretion of pigeon crop milk is different in the process of feeding the young, whether the metabolism and formation of pigeon milk use the same mechanism is a very interesting scientific question. However, the metabolic dynamics and underlying genetic mechanisms involved in the formation of pigeon crop milk remain unclear, particularly during the incubation-feeding reproductive cycle. In this study, we integrated lactation-associated metabolism and transcriptome data from the crop tissues of both male and female pigeons during the brooding and feeding stages. We mapped the changes in metabolites related to milk formation in the crop tissues during these stages. Through metabolome profiling, we identified 1413 metabolites among 18 crop tissues. During the breeding cycles, the concentrations of estrone, L-ergothioneine, and L-histidine exhibited the most dynamic changes in females. In contrast, estrone, L-anserine, 1-methylhistidine, homovanillate, oxidized glutathione, and reducing glutathione showed the most dynamic changes in males. Gender-specific differences were observed in the metabolome, with several metabolites significantly differing between males and females, many of which were correlated with cytokine binding, immunity, and cytochrome P450 activity. Using this dataset, we constructed complex regulatory networks, enabling us to identify important metabolites and key genes involved in regulating the formation of pigeon milk in male and female pigeons, respectively. Additionally, we investigated gender-associated differences in the crop metabolites of pigeons. Our study revealed differences in the modulation of pigeon crop milk metabolism between males and females and shed light on the potential functions of male and female pigeon milk in the growth, development, and immunity of young pigeons, an area that has not been previously explored. In conclusion, our results provide new insights into the metabolic regulation of pigeon crop milk formation during the brooding and breeding stages. Furthermore, our findings lay the foundation for the accurate development of artificial pigeon milk.
Collapse
Affiliation(s)
- Yuting Fu
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
- Waterfowl Healthy Breeding Engineering Research Center, Guangdong Higher Education Institute, Guangzhou 510225, China
| | - Yan Song
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
- Waterfowl Healthy Breeding Engineering Research Center, Guangdong Higher Education Institute, Guangzhou 510225, China
| | - Danli Jiang
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
- Waterfowl Healthy Breeding Engineering Research Center, Guangdong Higher Education Institute, Guangzhou 510225, China
| | - Jianqiu Pan
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
- Waterfowl Healthy Breeding Engineering Research Center, Guangdong Higher Education Institute, Guangzhou 510225, China
| | - Wanyan Li
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
- Waterfowl Healthy Breeding Engineering Research Center, Guangdong Higher Education Institute, Guangzhou 510225, China
| | - Xumeng Zhang
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
- Waterfowl Healthy Breeding Engineering Research Center, Guangdong Higher Education Institute, Guangzhou 510225, China
| | - Wenbin Chen
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Yunbo Tian
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
- Waterfowl Healthy Breeding Engineering Research Center, Guangdong Higher Education Institute, Guangzhou 510225, China
| | - Xu Shen
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
- Waterfowl Healthy Breeding Engineering Research Center, Guangdong Higher Education Institute, Guangzhou 510225, China
| | - Yunmao Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510225, China; (Y.F.); (Y.S.); (D.J.); (J.P.); (W.L.); (X.Z.); (W.C.); (Y.T.)
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
- Waterfowl Healthy Breeding Engineering Research Center, Guangdong Higher Education Institute, Guangzhou 510225, China
| |
Collapse
|
218
|
Cui Z, Amevor FK, Zhao X, Mou C, Pang J, Peng X, Liu A, Lan X, Liu L. Potential therapeutic effects of milk-derived exosomes on intestinal diseases. J Nanobiotechnology 2023; 21:496. [PMID: 38115131 PMCID: PMC10731872 DOI: 10.1186/s12951-023-02176-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/25/2023] [Indexed: 12/21/2023] Open
Abstract
Exosomes are extracellular vesicles with the diameter of 30 ~ 150 nm, and are widely involved in intercellular communication, disease diagnosis and drug delivery carriers for targeted disease therapy. Therapeutic application of exosomes as drug carriers is limited due to the lack of sources and methods for obtaining adequate exosomes. Milk contains abundant exosomes, several studies have shown that milk-derived exosomes play crucial roles in preventing and treating intestinal diseases. In this review, we summarized the biogenesis, secretion and structure, current novel methods used for the extraction and identification of exosomes, as well as discussed the role of milk-derived exosomes in treating intestinal diseases, such as inflammatory bowel disease, necrotizing enterocolitis, colorectal cancer, and intestinal ischemia and reperfusion injury by regulating intestinal immune homeostasis, restoring gut microbiota composition and improving intestinal structure and integrity, alleviating conditions such as oxidative stress, cell apoptosis and inflammation, and reducing mitochondrial reactive oxygen species (ROS) and lysosome accumulation in both humans and animals. In addition, we discussed future prospects for the standardization of milk exosome production platform to obtain higher concentration and purity, and complete exosomes derived from milk. Several in vivo clinical studies are needed to establish milk-derived exosomes as an effective and efficient drug delivery system, and promote its application in the treatment of various diseases in both humans and animals.
Collapse
Affiliation(s)
- Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, P. R. China
| | - Xingtao Zhao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Sichuan, P. R. China
| | - Chunyan Mou
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Jiaman Pang
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Xie Peng
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Anfang Liu
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Xi Lan
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China.
| | - Lingbin Liu
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China.
- College of Animal Science and Technology, Chongqing Key Laboratory of Forage & Herbivore, Chongqing Engineering Research Center for Herbivores Resource Protection and Utilization, Southwest University, Beibei, Chongqing, 400715, P. R. China.
| |
Collapse
|
219
|
Zhang Q, Luo Y, Peng L, Rong X, Liu Y, Li J, Luo J. Ferroptosis in cardiovascular diseases: role and mechanism. Cell Biosci 2023; 13:226. [PMID: 38102663 PMCID: PMC10724928 DOI: 10.1186/s13578-023-01169-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023] Open
Abstract
In multicellular organisms, regulatory cell death is a crucial aspect of growth and development. Ferroptosis, which was postulated roughly ten years ago, is a mode of cell death that differs from apoptosis, autophagy, and pyrodeath. This distinct pattern of cell death is triggered by an imbalance between oxidants and antioxidants and strongly associated with the metabolism of iron, lipids, amino acids, and glutathione. A growing body of research has implicated ferroptosis in the incidence and progression of many organ traumas and degenerative diseases. Recently, ferroptosis has gained attention as a crucial regulatory mechanism underlying the initiation and development of a variety of cardiovascular diseases, including myocardial ischemia/reperfusion injury, cardiomyopathy, arrhythmia, chemotherapy, and Corona Virus-2-induced cardiac injury. Pharmacological therapies that inhibit ferroptosis have great potential for the management of cardiovascular disorders. This review discusses the prevalence and regulatory mechanisms of ferroptosis, effect of ferroptosis on the immune system, significance of ferroptosis in cardiovascular diseases, and potential therapeutic value of regulating ferroptosis in a variety of heart diseases.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhao Luo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lin Peng
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xi Rong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yingxue Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiafu Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Collaborative Innovation Centre for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.
| | - Jing Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Collaborative Innovation Centre for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.
| |
Collapse
|
220
|
Li C, Wu Y, Chen K, Chen R, Xu S, Yang B, Lian Z, Wang X, Wang K, Xie H, Zheng S, Liu Z, Wang D, Xu X. Gp78 deficiency in hepatocytes alleviates hepatic ischemia-reperfusion injury via suppressing ACSL4-mediated ferroptosis. Cell Death Dis 2023; 14:810. [PMID: 38065978 PMCID: PMC10709349 DOI: 10.1038/s41419-023-06294-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023]
Abstract
Ferroptosis, which is driven by iron-dependent lipid peroxidation, plays an essential role in liver ischemia-reperfusion injury (IRI) during liver transplantation (LT). Gp78, an E3 ligase, has been implicated in lipid metabolism and inflammation. However, its role in liver IRI and ferroptosis remains unknown. Here, hepatocyte-specific gp78 knockout (HKO) or overexpressed (OE) mice were generated to examine the effect of gp78 on liver IRI, and a multi-omics approach (transcriptomics, proteomics, and metabolomics) was performed to explore the potential mechanism. Gp78 expression decreased after reperfusion in LT patients and mice with IRI, and gp78 expression was positively correlated with liver damage. Gp78 absence from hepatocytes alleviated liver damage in mice with IRI, ameliorating inflammation. However, mice with hepatic gp78 overexpression showed the opposite phenotype. Mechanistically, gp78 overexpression disturbed lipid homeostasis, remodeling polyunsaturated fatty acid (PUFA) metabolism, causing oxidized lipids accumulation and ferroptosis, partly by promoting ACSL4 expression. Chemical inhibition of ferroptosis or ACSL4 abrogated the effects of gp78 on ferroptosis and liver IRI. Our findings reveal a role of gp78 in liver IRI pathogenesis and uncover a mechanism by which gp78 promotes hepatocyte ferroptosis by ACSL4, suggesting the gp78-ACSL4 axis as a feasible target for the treatment of IRI-associated liver damage.
Collapse
Affiliation(s)
- Changbiao Li
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Yichao Wu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Kangchen Chen
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Ronggao Chen
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shengjun Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Beng Yang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhengxing Lian
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Xiaodong Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Kai Wang
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Haiyang Xie
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shusen Zheng
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou, 311112, China
| | - Zhikun Liu
- Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China.
| | - Di Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China.
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China.
| |
Collapse
|
221
|
An F, Zhang J, Gao P, Xiao Z, Chang W, Song J, Wang Y, Ma H, Zhang R, Chen Z, Yan C. New insight of the pathogenesis in osteoarthritis: the intricate interplay of ferroptosis and autophagy mediated by mitophagy/chaperone-mediated autophagy. Front Cell Dev Biol 2023; 11:1297024. [PMID: 38143922 PMCID: PMC10748422 DOI: 10.3389/fcell.2023.1297024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
Ferroptosis, characterized by iron accumulation and lipid peroxidation, is a form of iron-driven cell death. Mitophagy is a type of selective autophagy, where degradation of damaged mitochondria is the key mechanism for maintaining mitochondrial homeostasis. Additionally, Chaperone-mediated autophagy (CMA) is a biological process that transports individual cytoplasmic proteins to lysosomes for degradation through companion molecules such as heat shock proteins. Research has demonstrated the involvement of ferroptosis, mitophagy, and CMA in the pathological progression of Osteoarthritis (OA). Furthermore, research has indicated a significant correlation between alterations in the expression of reactive oxygen species (ROS), adenosine monophosphate (AMP)-activated protein kinase (AMPK), and hypoxia-inducible factors (HIFs) and the occurrence of OA, particularly in relation to ferroptosis and mitophagy. In light of these findings, our study aims to assess the regulatory functions of ferroptosis and mitophagy/CMA in the pathogenesis of OA. Additionally, we propose a mechanism of crosstalk between ferroptosis and mitophagy, while also examining potential pharmacological interventions for targeted therapy in OA. Ultimately, our research endeavors to offer novel insights and directions for the prevention and treatment of OA.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhipan Xiao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Weirong Chang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jiayi Song
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yujie Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Haizhen Ma
- Teaching Department of Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Rui Zhang
- Teaching Department of Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhendong Chen
- Teaching Department of Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
222
|
Huang J, Xu Z, Jiao J, Li Z, Li S, Liu Y, Li Z, Qu G, Wu J, Zhao Y, Chen K, Li J, Pan Y, Wu X, Ren J. Microfluidic intestinal organoid-on-a-chip uncovers therapeutic targets by recapitulating oxygen dynamics of intestinal IR injury. Bioact Mater 2023; 30:1-14. [PMID: 37534235 PMCID: PMC10391666 DOI: 10.1016/j.bioactmat.2023.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/11/2023] [Accepted: 07/01/2023] [Indexed: 08/04/2023] Open
Abstract
Increasing evidence demonstrates that mammals have different reactions to hypoxia with varied oxygen dynamic patterns. It takes ∼24 h for tri-gas incubator to achieve steady cell hypoxia, which fails to recapitulate ultrafast oxygen dynamics of intestinal ischemia/reperfusion (IR) injury. Inspired from the structure of native intestinal villi, we engineered an intestinal organoid chip embedded with engineered artificial microvessels based on co-axial microfluidic technology by using pH-responsive ZIF-8/sodium alginate scaffold. The chip was featured on: (i) eight times the oxygen exchange efficiency compared with the conventional device, tri-gas incubator, (ii) implantation of intestinal organoid reproducing all types of intestinal epithelial cells, and (iii) bio-responsiveness to hypoxia and reoxygenation (HR) by presenting metabolism disorder, inflammatory reaction, and cell apoptosis. Strikingly, it was found for the first time that Olfactomedin 4 (Olfm4) was the most significantly down-regulated gene under a rapid HR condition by sequencing the RNA from the organoids. Mechanistically, OLFM4 played protective functions on HR-induced cell inflammation and tissue damage by inhibiting the NF-kappa B signaling activation, thus it could be used as a therapeutic target. Altogether, this study overcomes the issue of mismatched oxygen dynamics between in vitro and in vivo, and sets an example of next-generation multisystem-interactive organoid chip for finding precise therapeutic targets of IR injury.
Collapse
Affiliation(s)
- Jinjian Huang
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ziyan Xu
- School of Medicine, Nanjing University, Nanjing, 210093, China
| | - Jiao Jiao
- Department of Rehabilitation, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zongan Li
- Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing, NARI School of Electrical and Automation Engineering, Nanjing Normal University, Nanjing, 210042, China
| | - Sicheng Li
- School of Medicine, Nanjing University, Nanjing, 210093, China
| | - Ye Liu
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ze Li
- School of Medicine, Nanjing University, Nanjing, 210093, China
| | - Guiwen Qu
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jie Wu
- General Clinical Research Center, Nanjing Benq Hospital, Nanjing Medical University, Nanjing, 210019, China
| | - Yun Zhao
- General Clinical Research Center, Nanjing Benq Hospital, Nanjing Medical University, Nanjing, 210019, China
| | - Kang Chen
- School of Medicine, Nanjing University, Nanjing, 210093, China
| | - Jieshou Li
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yichang Pan
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
- School of Medicine, Nanjing University, Nanjing, 210093, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
- School of Medicine, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
223
|
Abstract
Ferroptosis is a distinctive form of iron-dependent necrotic cell death, characterized by excessive lipid peroxidation on cellular membranes and compromised cellular antioxidant defenses. Multiple metabolic pathways, including iron and lipid metabolism, as well as antioxidant systems, contribute to the execution of ferroptosis. The gut microbiota exerts regulatory effects on ferroptosis through its microbial composition, biological functions, and metabolites. Notably, most pathogenic bacteria tend to promote ferroptosis, thereby inducing or exacerbating diseases, while most probiotics have been shown to protect against cell death. Given microbiota colonization in the gut, an intimate association is found between intestinal diseases and microbiota. This review consolidates the essential aspects of ferroptotic processes, emphasizing key molecules and delineating the intricate interplay between gut microbiota and ferroptosis. Moreover, this review underscores the potential utility of gut microbiota modulation in regulating ferroptosis for the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Ting Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
| |
Collapse
|
224
|
Wang Y, Li B, Liu G, Han Q, Diao Y, Liu J. Corilagin attenuates intestinal ischemia/reperfusion injury in mice by inhibiting ferritinophagy-mediated ferroptosis through disrupting NCOA4-ferritin interaction. Life Sci 2023; 334:122176. [PMID: 37858718 DOI: 10.1016/j.lfs.2023.122176] [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: 08/24/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
AIMS Intestinal ischemia reperfusion (II/R) is a common clinical emergency. Ferroptosis is reported to play a role in II/R injury. Our previous studies revealed that corilagin significantly attenuates intestinal ischemia/reperfusion injuries. However, the underlying molecular mechanism is unclear and requires further study. MATERIALS AND METHODS DAO, GSSG/T-GSH, MDA, and Fe2+ were measured by assay kits, 4-HNE was assessed by IHC, and 15-LOX was measured by ELISA. Mitochondrial damage was observed by TEM and cellular oxidation levels were detected by C11-BODIPY 581/591 and DHE probes. LC3, p62, Beclin1, ACSL4, GPX4, NCOA4, and ferritin expression were examined by WB in vivo and in vitro. IF, co-IF, q-PCR, and constructed NCOA4-knock-down IEC-6 cells were used to evaluate the role of NCOA4 in the effect of corilagin against II/R injury. Temporal and nucleoplasmic variations with or without corilagin were observed by WB. Co-IP and molecular docking were used to investigate the NCOA4-ferritin interaction. KEY FINDINGS Corilagin attenuated II/R-induced ferroptosis both in vitro and in vivo. Further study revealed that the anti-ferroptosis bioactivity of corilagin might be due to the modulation of iron homeostasis via inhibition of ferritinophagy in an NCOA4-dependent manner. SIGNIFICANCE Corilagin might be a potential therapeutic agent for II/R-induced tissue injury.
Collapse
Affiliation(s)
- Yunxiang Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Bin Li
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian 116044, China
| | - Guanting Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Qipeng Han
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian 116044, China.
| | - Jing Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian 116044, China.
| |
Collapse
|
225
|
Wu H, Li H, Huo H, Li X, Zhu H, Zhao L, Liao J, Tang Z, Guo J. Effects of terbuthylazine on myocardial oxidative stress and ferroptosis via Nrf2/HO-1 signaling pathway in broilers. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 197:105698. [PMID: 38072553 DOI: 10.1016/j.pestbp.2023.105698] [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: 10/12/2023] [Revised: 11/04/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023]
Abstract
Terbuthylazine (TBA) is one of the most commonly used and effective herbicides. However, due to its affinity for soil organic matter and water solubility, TBA can lead to biological health concerns. This study exposed broilers to TBA (0 mg/kg bw, 0.4 mg/kg bw, 4 mg/kg bw) for 28 days. The results showed significant pathological damage in broiler myocardial tissue, such as widening of the interstitial space, rupture of muscle fibers, and deposition of myocardial collagen fibers. In addition, Under the 0.4 mg/kg bw TBA exposure, myocardial oxidative stress was observed in broilers, which was accompanied by the activation of Nrf2/HO-1 pathway and the increased protein and mRNA levels of NQO1, NOX2 and SOD2 antioxidant enzymes. However, Nrf2/HO-1 protein and mRNA levels were reversed at 4 mg/kg bw TBA exposure. Meanwhile, the Nrf2/HO-1 mediated antioxidant defense was impaired. In contrast with the low dose, the protein and gene expression levels of NQO1, NOX2, and SOD2 were reduced in 4 mg/kg bw TBA group. The expression of GPX4 and SLC7A11 was significantly downregulated at both protein and mRNA levels. Beyond that, ACSL4 expression was significantly up-regulated, and the protein result was consistent with the mRNA expression, demonstrating the occurrence of ferroptosis. In general, TBA exposure activated the Nrf2/HO-1 pathway, resulting in ferroptosis. This study links ferroptosis to the Nrf2/HO-1 pathway, providing new insights into the potential role of TBA in myocardial toxicity.
Collapse
Affiliation(s)
- Haitong Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Haoye Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Haihua Huo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xinrun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Heyun Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Lijiao Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jianying Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
226
|
Wan Z, zhang Y, Lv J, Yuan Y, Guo W, Leng Y. Exosomes derived from bone marrow mesenchymal stem cells regulate pyroptosis via the miR-143-3p/myeloid differentiation factor 88 axis to ameliorate intestinal ischemia-reperfusion injury. Bioengineered 2023; 14:2253414. [PMID: 37674357 PMCID: PMC10486297 DOI: 10.1080/21655979.2023.2253414] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/02/2022] [Accepted: 08/20/2022] [Indexed: 09/08/2023] Open
Abstract
Intestinal ischemia-reperfusion (I/R) injury is a condition in which tissue injury is aggravated after ischemia due to recovery of blood supply. Bone marrow mesenchymal stem cell-derived exosome (BMSC-exo) showed a protective effect on I/R injury. This study aimed to investigate the possible mechanisms by which BMSC-exos ameliorate intestinal I/R injury. We isolated mouse BMSC-exos by super-centrifugation and found that they effectively increased cell viability in a cell model, alleviated intestinal barrier injury in a mouse model, and downregulated the expression of inflammatory cytokines and pyroptosis-related proteins, suggesting that BMSC-exos may alleviate intestinal I/R injury in vitro and in vivo by regulating pyroptosis. We identified miR-143-3p as a differentially expressed miRNA by microarray sequencing. Bioinformatic analysis predicted a binding site between miR-143-3p and myeloid differentiation factor 88 (MyD88); a dual-luciferase reporter assay confirmed that miR-143-3p could directly regulate the expression of MyD88. Our findings suggest that miR-143-3p regulates pyroptosis by regulating NOD-like receptor thermal protein domain associated protein 3 (NLRP3) through the toll-like receptor (TLR)-4/MyD88/nuclear factor kappa-B (NF-кB) pathway. This study describes a potential strategy for the treatment of intestinal I/R injury using BMSC-exos that act by regulating pyroptosis through the miR-143-3p mediated TLR4/MyD88/NF-кB pathway.
Collapse
Affiliation(s)
- Zhanhai Wan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Yan zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Jipeng Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Yuan Yuan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Wenwen Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Yufang Leng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| |
Collapse
|
227
|
Shen J, Qian M, Wu M, Tang J, Gong Y, Li J, Ji J, Dang B. Rosiglitazone inhibits acyl-CoA synthetase long-chain family number 4 and improves secondary brain injury in a rat model of surgical brain injury. Clin Exp Pharmacol Physiol 2023; 50:927-935. [PMID: 37675456 DOI: 10.1111/1440-1681.13815] [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: 11/21/2022] [Revised: 08/02/2023] [Accepted: 08/05/2023] [Indexed: 09/08/2023]
Abstract
Ferroptosis is a recently discovered non-apoptotic form of cellular death. Acyl-CoA synthetase long-chain family number 4 (ACSL4) is necessary for iron-dependent cellular death, and reactive oxygen species (ROS) produced by ACSL4 are the executioners of ferroptosis. Rosiglitazone improves ferroptosis by inhibiting ACSL4. There is no research indicating whether ACSL4 plays a role in cell death after surgical brain injury (SBI). This study aimed to investigate the role of ACSL4 in SBI via the ferroptosis pathway. Ninety male Sprague-Dawley rats were examined using a model of SBI. Subsequently, the inhibitory effect of rosiglitazone on ACSL4 was assessed via western blot, real-time polymerase chain reaction (PCR), immunofluorescence, fluoro-jade C staining, Perl's staining, ROS assay, and neurological scoring. The results showed that compared with the Sham group, the protein levels of ACSL4 and transferrin were significantly increased after SBI. Administration of rosiglitazone significantly reduced neuronal necrosis, iron deposition, brain water content and ROS in brain tissue and ameliorated neurological deficits at 48 h after SBI, which was concomitant with decreased transferrin expression. These findings demonstrate that SBI-induced upregulation of ACSL4 may be partly mediated by the ferroptosis pathway, which can be reversed by rosiglitazone administration.
Collapse
Affiliation(s)
- Jinchao Shen
- Departments of Anesthesiology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| | - Min Qian
- Departments of Anesthesiology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| | - Muyao Wu
- Departments of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| | - Jiafeng Tang
- Departments of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| | - Yating Gong
- Departments of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| | - Jie Li
- Departments of Intensive Care Unit, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, No.77 Changan Southern Road, Jiangsu Province, China
| | - Jinfen Ji
- Departments of Anesthesiology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| | - Baoqi Dang
- Departments of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| |
Collapse
|
228
|
Liu X, Ren M, Zhang A, Huang C, Wang J. Nrf2 attenuates oxidative stress to mediate the protective effect of ciprofol against cerebral ischemia-reperfusion injury. Funct Integr Genomics 2023; 23:345. [PMID: 37996761 DOI: 10.1007/s10142-023-01273-z] [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: 09/03/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
Neuroinflammation and oxidative stress damage are involved in the pathogenesis of cerebral ischemia-reperfusion injury (CIRI). Ferroptosis emerged as a new player in the regulation of lipid peroxidation processes. This study aimed at exploring the potential involvement of ciprofol on ferroptosis-associated CIRI and subsequent neurological deficits in the mouse model of transient cerebral ischemia and reperfusion. Cerebral ischemia was built in male C57BL/6 J wild-type (WT) and Nrf2-knockout (Nrf2 KO) mice in the manner of middle cerebral artery occlusion (MCAO) followed by reperfusion. Ciprofol improved autonomic behavior, alleviated reactive oxygen species output and ferroptosis-induced neuronal death by nucleus transportation of NFE2 like BZIP transcription factor 2 (Nrf2) and the promotion of heme oxygenase 1 (Ho-1), solute carrier family 7 member 11 (SLC7A11/xCT), and glutathione peroxidase 4 (GPX4). Additionally, ciprofol improved neurological scores and reduced infarct volume, brain water content, and necrotic neurons. Cerebral blood flow in MCAO-treated mice was also improved. Furthermore, absence of Nrf2 abrogated the neuroprotective actions of ciprofol on antioxidant capacity and sensitized neurons to oxidative stress damage. In vitro, the primary-cultured cortical neurons from mice were pre-treated with oxygen-glucose deprivation/reperfusion (OGD/R), followed by ciprofol administration. Ciprofol effectively reversed OGD/R-induced ferroptosis and accelerated transcription of GPX4 and xCT. In conclusion, we investigated the ciprofol-induced inhibition effect of ferroptosis-sheltered neurons from lipid preoxidation in the pathogenesis of CIRI via Nrf2-xCT-GPX4 signaling pathway.
Collapse
Affiliation(s)
- Xia Liu
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Zhejiang Province, Ningbo, 315000, China
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province, Wenzhou, 325000, China
| | - Miao Ren
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province, Wenzhou, 325000, China
| | - Anqi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province, Wenzhou, 325000, China
| | - Changshun Huang
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Zhejiang Province, Ningbo, 315000, China.
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province, Wenzhou, 325000, China.
| |
Collapse
|
229
|
Gao S, Sun C, Kong J. Vitamin D Attenuates Ulcerative Colitis by Inhibiting ACSL4-Mediated Ferroptosis. Nutrients 2023; 15:4845. [PMID: 38004239 PMCID: PMC10675831 DOI: 10.3390/nu15224845] [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: 10/07/2023] [Revised: 10/19/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND With environmental and lifestyle changes, recent epidemiological studies have shown that the prevalence of Ulcerative Colitis (UC) is on the rise, while treatment options are limited. There is an urgent need to explore the underlying mechanisms of vitamin D (VD) as an effective treatment. METHODS Dextran sulfate sodium-induced mice and lipopolysaccharide-induced HCT116 cells were used to establish the classic UC models in vivo and in vitro, respectively. Typical symbols of inflammation (IL-6, COX-2), oxidative stress (MDA, MPO, GSH), and ferroptosis (ACSL4, GPX4, SLC7A11, and Iron) were analyzed by Western blot, Immunohistochemistry, RT-PCR, and relative assay kits. The inflammation factors and oxidative stress injury of cells transfected with ACSL4+/+ plasmids were tested by Western blot, MDA, and MPO methods. RESULTS Vitamin D attenuated the levels of COX-2, IL-6, Iron, MDA, and MPO and improved SOD1 and GSH contents in DSS + VD and LPS + VD groups, compared with model groups. Ferrostatin-1 (Fer-1) could relieve the levels of COX-2, IL-6, Iron, MDA, and MPO while increasing the contents of SOD1 and GSH in DSS + Fer-1 and LPS + Fer-1 compared to model groups. VD downregulated the expression of ACSL4 and upregulated GPX4 in tissues and cells. After transfected with ACSL4+/+ plasmids, we found VD's role of downregulating inflammation and oxidative stress was relieved. CONCLUSIONS Vitamin D can relieve UC by inhibiting ferroptosis both in mice and in cells through the negative regulation of ACSL4, providing new insight into the therapeutic function of VD on UC.
Collapse
Affiliation(s)
- Shuo Gao
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Can Sun
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Juan Kong
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
230
|
Ren K, Pei J, Guo Y, Jiao Y, Xing H, Xie Y, Yang Y, Feng Q, Yang J. Regulated necrosis pathways: a potential target for ischemic stroke. BURNS & TRAUMA 2023; 11:tkad016. [PMID: 38026442 PMCID: PMC10656754 DOI: 10.1093/burnst/tkad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/24/2022] [Indexed: 12/01/2023]
Abstract
Globally, ischemic stroke causes millions of deaths per year. The outcomes of ischemic stroke are largely determined by the amount of ischemia-related and reperfusion-related neuronal death in the infarct region. In the infarct region, cell injuries follow either the regulated pathway involving precise signaling cascades, such as apoptosis and autophagy, or the nonregulated pathway, which is uncontrolled by any molecularly defined effector mechanisms such as necrosis. However, numerous studies have recently found that a certain type of necrosis can be regulated and potentially modified by drugs and is nonapoptotic; this type of necrosis is referred to as regulated necrosis. Depending on the signaling pathway, various elements of regulated necrosis contribute to the development of ischemic stroke, such as necroptosis, pyroptosis, ferroptosis, pathanatos, mitochondrial permeability transition pore-mediated necrosis and oncosis. In this review, we aim to summarize the underlying molecular mechanisms of regulated necrosis in ischemic stroke and explore the crosstalk and interplay among the diverse types of regulated necrosis. We believe that targeting these regulated necrosis pathways both pharmacologically and genetically in ischemia-induced neuronal death and protection could be an efficient strategy to increase neuronal survival and regeneration in ischemic stroke.
Collapse
Affiliation(s)
- Kaidi Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Jinyan Pei
- Quality Management Department, Henan No. 3 Provincial People’s Hospital, Henan No. 3 Provincial People’s Hospital, Zhengzhou 450052, China
| | - Yuanyuan Guo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yuxue Jiao
- Quality Management Department, Henan No. 3 Provincial People’s Hospital, Henan No. 3 Provincial People’s Hospital, Zhengzhou 450052, China
| | - Han Xing
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yang Yang
- Research Center for Clinical System Biology, Translational Medicine Center, No. 1 Jianshe Dong Road, ErQi District, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qi Feng
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
| | - Jing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
231
|
Wang X, Li M, Diao K, Wang Y, Chen H, Zhao Z, Li Y, Jia X, Wang H, Zheng F, Xia Z, Han L, Zhang M. Deferoxamine attenuates visual impairment in retinal ischemia‒reperfusion via inhibiting ferroptosis. Sci Rep 2023; 13:20145. [PMID: 37978208 PMCID: PMC10656451 DOI: 10.1038/s41598-023-46104-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023] Open
Abstract
Retinal ischemia‒reperfusion (I/R) injury can cause significant damage to human retinal neurons, greatly compromising their functions. Existing interventions have been proven to have little effect. Ferroptosis is a newly discovered type of programmed cell death that has been found to be involved in the process of ischemia‒reperfusion in multiple organs throughout the body. Studies have shown that it is also present in retinal ischemia‒reperfusion injury. A rat model of retinal ischemia‒reperfusion injury was constructed and treated with deferoxamine. In this study, we found the accumulation of Fe2+, reactive oxygen species (ROS), malondialdehyde (MDA), and the consumption of glutathione (GSH) via ELISA testing; increased expression of transferrin; and decreased expression of ferritin, SLC7A11, and GPX4 via Western blotting (WB) and real-time PCR testing. Structural signs of ferroptosis (mitochondrial shrinkage) were observed across multiple cell types, including retinal ganglion cells (RGCs), photoreceptor cells, and pigment epithelial cells. Changes in visual function were detected by F-VEP and ERG. The results showed that iron and oxidative stress were increased in the retinal ischemia‒reperfusion injury model, resulting in ferroptosis and tissue damage. Deferoxamine protects the structural and functional soundness of the retina by inhibiting ferroptosis through the simultaneous inhibition of hemochromatosis, the initiation of transferrin, and the degradation of ferritin and activating the antioxidant capacity of the System Xc-GSH-GPX4 pathway.
Collapse
Affiliation(s)
- Xiaoxuan Wang
- Department of Ophthalmology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Mingran Li
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Ke Diao
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Yan Wang
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Hong Chen
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Ziqi Zhao
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Yuan Li
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Xin Jia
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Hao Wang
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Fangyuan Zheng
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Zihan Xia
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Longhui Han
- Department of Ophthalmology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China
| | - Minglian Zhang
- Department of Ophthalmology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
- Hebei Eye Hospital, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Clinical Research Center for Eye Diseases, Xingtai, 054000, Hebei, China.
| |
Collapse
|
232
|
Hou M, Chen F, He Y, Tan Z, Han X, Shi Y, Xu Y, Leng Y. Dexmedetomidine against intestinal ischemia/reperfusion injury: A systematic review and meta-analysis of preclinical studies. Eur J Pharmacol 2023; 959:176090. [PMID: 37778612 DOI: 10.1016/j.ejphar.2023.176090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 09/14/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Intestinal ischemia/reperfusion injury (IRI) is a multifactorial, complex pathophysiological process in clinical settings. In recent years, intestinal IRI has received increasing attention due to increased morbidity and mortality. To date, there are no effective treatments. Dexmedetomidine (DEX), a highly selective α2-adrenergic receptor agonist, has been demonstrated to be effective against intestinal IRI. In this systematic review and meta-analysis, we evaluated the efficacy and potential mechanisms of DEX as a treatment for intestinal IRI in animal models. METHODS Five databases (PubMed, Embase, Web of Science, Cochrane Library, and Scopus) were searched until March 15, 2023. Using the SYRCLE risk bias tool, we assessed methodological quality. Statistical analysis was conducted using STATA 12 and R 4.2.2. We analyzed the related outcomes (mucosa damage-related indicators; inflammation-relevant markers, oxidative stress markers) relied on the fixed or random-effects models. RESULTS There were 15 articles including 18 studies included, and 309 animals were involved in the studies. Compared to the model groups, DEX improved intestinal IRI. DEX decreased Chiu's score and serum diamine oxidase (DAO) level. DEX reduced the level of inflammation-relevant markers (interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α). DEX also improved oxidative stress (decreased malondialdehyde (MDA), increased superoxide dismutase (SOD)). CONCLUSIONS DEX's effectiveness in ameliorating intestinal IRI has been demonstrated in animal models. Antioxidation, anti-inflammation, anti-apoptotic, anti-pyroptosis, anti-ferroptosis, enhancing mitophagy, reshaping the gut microbiota, and gut barrier protection are possible mechanisms. However, in light of the heterogeneity and methodological quality of these studies, further well-designed preclinical studies are warranted before clinical implication.
Collapse
Affiliation(s)
- Min Hou
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| | - Feng Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| | - Yao He
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| | - Zhiguo Tan
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| | - Xuena Han
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| | - Yajing Shi
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| | - Yunpeng Xu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| | - Yufang Leng
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China; Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
233
|
Huang Y, Chen L, Xiong B, Lu G, Chen C, Liu J. Integrating multiple microarray datasets to explore the significance of ferroptosis regulators in the diagnosis and subtype classification of osteoarthritis. Medicine (Baltimore) 2023; 102:e35917. [PMID: 37960823 PMCID: PMC10637513 DOI: 10.1097/md.0000000000035917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease that reduces quality of life for patients. Ferroptosis plays a significant role in OA. However, its underlying mechanism remains unclear. In this study, we integrated 7 OA synovial datasets from the GEO database to screen for significant ferroptosis-related genes. The top 5 ferroptosis regulators were used to construct nomogram models to predict OA prevalence. Consensus clustering was applied to classify OA patients into different ferroptosis patterns based on significant ferroptosis-related genes. Subsequently, an immune cell infiltration study was performed to investigate the relationship between the significant ferroptosis regulators and immune cells. As a result, we screened 11 ferroptosis-related genes in OA patients. Five candidate ferroptosis regulators (SLC7A11, ALOX5, SLC1A5, GOT1, and GSS) were used to predict OA risk. The nomogram model based on these 5 genes is important for assessing the occurrence of OA. Consensus clustering analysis showed that OA patients could be classified into 2 ferroptosis patterns (Clusters A and B). Immune cell infiltration levels were higher in Cluster B than in Cluster A. Two subtypes, gene Clusters A and B, were classified according to the expression of ferroptosis-related DEGs among the ferroptosis patterns. Cluster A and gene Cluster A had higher ferroptosis scores than Cluster B or gene Cluster B, whereas the expression levels of the proinflammatory cytokines interleukin (IL)-1β, tumor necrosis factor, IL-6, IL-18, and IL-10 were higher in Cluster B or gene Cluster B than those in Cluster A or gene Cluster A. Different subtypes of ferroptosis play critical roles in OA. Furthermore, immunotherapy strategies for OA treatment may be guided by our study on ferroptosis patterns.
Collapse
Affiliation(s)
- Yue Huang
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - Lihua Chen
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - Bo Xiong
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - GuanYu Lu
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - Cai Chen
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - JinFu Liu
- Department of Orthopedics and Traumatology, Xianhu District, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Guangxi, China
| |
Collapse
|
234
|
Ding K, Liu C, Li L, Yang M, Jiang N, Luo S, Sun L. Acyl-CoA synthase ACSL4: an essential target in ferroptosis and fatty acid metabolism. Chin Med J (Engl) 2023; 136:2521-2537. [PMID: 37442770 PMCID: PMC10617883 DOI: 10.1097/cm9.0000000000002533] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Indexed: 07/15/2023] Open
Abstract
ABSTRACT Long-chain acyl-coenzyme A (CoA) synthase 4 (ACSL4) is an enzyme that esterifies CoA into specific polyunsaturated fatty acids, such as arachidonic acid and adrenic acid. Based on accumulated evidence, the ACSL4-catalyzed biosynthesis of arachidonoyl-CoA contributes to the execution of ferroptosis by triggering phospholipid peroxidation. Ferroptosis is a type of programmed cell death caused by iron-dependent peroxidation of lipids; ACSL4 and glutathione peroxidase 4 positively and negatively regulate ferroptosis, respectively. In addition, ACSL4 is an essential regulator of fatty acid (FA) metabolism. ACSL4 remodels the phospholipid composition of cell membranes, regulates steroidogenesis, and balances eicosanoid biosynthesis. In addition, ACSL4-mediated metabolic reprogramming and antitumor immunity have attracted much attention in cancer biology. Because it facilitates the cross-talk between ferroptosis and FA metabolism, ACSL4 is also a research hotspot in metabolic diseases and ischemia/reperfusion injuries. In this review, we focus on the structure, biological function, and unique role of ASCL4 in various human diseases. Finally, we propose that ACSL4 might be a potential therapeutic target.
Collapse
Affiliation(s)
- Kaiyue Ding
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410000, China
| | - Chongbin Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410000, China
| | - Li Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410000, China
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410000, China
| | - Na Jiang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410000, China
| | - Shilu Luo
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410000, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410000, China
| |
Collapse
|
235
|
Du L, Yang H, Ren Y, Ding Y, Xu Y, Zi X, Liu H, He P. Inhibition of LSD1 induces ferroptosis through the ATF4-xCT pathway and shows enhanced anti-tumor effects with ferroptosis inducers in NSCLC. Cell Death Dis 2023; 14:716. [PMID: 37923740 PMCID: PMC10624898 DOI: 10.1038/s41419-023-06238-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023]
Abstract
Lysine-specific demethylase 1 (LSD1) has been identified as an important epigenetic target, and recent advances in lung cancer therapy have highlighted the importance of targeting ferroptosis. However, the precise mechanisms by which LSD1 regulates ferroptosis remain elusive. In this study, we report that the inhibition of LSD1 induces ferroptosis by enhancing lipid peroxidation and reactive oxygen species (ROS) accumulation. Mechanistically, LSD1 inhibition downregulates the expression of activating transcription factor 4 (ATF4) through epigenetic modification of histone H3 lysine 9 dimethyl (H3K9me2), which sequentially inhibits the expression of the cystine-glutamate antiporter (xCT) and decreases glutathione (GSH) production. Furthermore, LSD1 inhibition transcriptionally upregulates the expression of transferrin receptor (TFRC) and acyl-CoA synthetase long chain family member 4 (ACSL4) by enhancing the binding of histone H3 lysine 4 dimethyl (H3K4me2) to their promoter sequences. Importantly, the combination of an LSD1 inhibitor and a ferroptosis inducer demonstrates an enhanced anti-tumor effect in a xenograft model of non-small cell lung cancer (NSCLC), surpassing the efficacy of either agent alone. These findings reveal new insights into the mechanisms by which LSD1 inhibition induces ferroptosis, offering potential guidance for the development of new strategies in the treatment of NSCLC.
Collapse
Affiliation(s)
- Linna Du
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Han Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yufei Ren
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yanli Ding
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yichao Xu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaolin Zi
- Departments of Urology and Pharmaceutical Sciences and Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, 92697, USA
| | - Hongmin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Pengxing He
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
236
|
Huang S, Liu Y, Wang C, Xiang W, Wang N, Peng L, Jiang X, Zhang X, Fu Z. Strategies for Cartilage Repair in Osteoarthritis Based on Diverse Mesenchymal Stem Cells-Derived Extracellular Vesicles. Orthop Surg 2023; 15:2749-2765. [PMID: 37620876 PMCID: PMC10622303 DOI: 10.1111/os.13848] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
Osteoarthritis (OA) causes disability and significant economic and social burden. Cartilage injury is one of the main pathological features of OA, and is often manifested by excessive chondrocyte death, inflammatory response, abnormal bone metabolism, imbalance of extracellular matrix (ECM) metabolism, and abnormal vascular or nerve growth. Regrettably, due to the avascular nature of cartilage, its capacity to repair is notably limited. Mesenchymal stem cells-derived extracellular vesicles (MSCs-EVs) play a pivotal role in intercellular communication, presenting promising potential not only as early diagnostic biomarkers in OA but also as efficacious therapeutic strategy. MSCs-EVs were confirmed to play a therapeutic role in the pathological process of cartilage injury mentioned above. This paper comprehensively provides the functions and mechanisms of MSCs-EVs in cartilage repair.
Collapse
Affiliation(s)
- Shanjun Huang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Yujiao Liu
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Chenglong Wang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Wei Xiang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Nianwu Wang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Li Peng
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Xuanang Jiang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiaomin Zhang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Zhijiang Fu
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
237
|
Deng X, Chu W, Zhang H, Peng Y. Nrf2 and Ferroptosis: A New Research Direction for Ischemic Stroke. Cell Mol Neurobiol 2023; 43:3885-3896. [PMID: 37728817 PMCID: PMC11407729 DOI: 10.1007/s10571-023-01411-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 09/04/2023] [Indexed: 09/21/2023]
Abstract
Ischemic stroke (IS) is one of the leading causes of death and morbidity worldwide. As a novel form of cell death, ferroptosis is an important mechanism of ischemic stroke. Nuclear factor E2-related factor 2 (Nrf2) is the primary regulator of cellular antioxidant response. In addition to alleviating ischemic stroke nerve damage by reducing oxidative stress, Nrf2 regulates genes associated with ferroptosis, suggesting that Nrf2 may inhibit ferroptosis after ischemic stroke. However, the specific pathway of Nrf2 on ferroptosis in the field of ischemic stroke remains unclear. Therefore, this paper provides a concise overview of the mechanisms underlying ferroptosis, with a particular focus on the regulatory role of Nrf2. The discussion highlights the potential connections between Nrf2 and the mitigation of oxidative stress, regulation of iron metabolism, modulation of the interplay between ferroptosis and inflammation, as well as apoptosis. This paper focuses on the specific pathway of Nrf2 regulation of ferroptosis after ischemic stroke, providing scientific research ideas for further research on the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Xiaoman Deng
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Wenming Chu
- Henan University of Chinese Medicine, Zhengzhou, 450000, Henan Province, China
| | - Hanrui Zhang
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Yongjun Peng
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
238
|
Chen X, Chen J, Miao C, Yin G, Zhang Z, Sun R, Ni S. Acetyl zingerone ameliorates osteoarthritis by inhibiting chondrocyte programmed cell death. Mol Med Rep 2023; 28:202. [PMID: 37711057 PMCID: PMC10540024 DOI: 10.3892/mmr.2023.13089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/16/2023] [Indexed: 09/16/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative disease that ultimately leads to joint deformity. The pathogenesis of OA is believed to involve abnormal chondrocyte death, with ferroptosis serving a key role in chondrocyte damage. The present study investigated whether acetyl zingerone (AZ), a newly identified antioxidant derived from curcumin, can alleviate the progression of OA. To investigate this, the present study performed various experiments, including crystal violet staining, flow cytometry, immunofluorescence and western blot analysis. In addition, dual validation was performed using in vivo and in vitro experiments; a mouse OA model was constructed for the in vivo experiments, and chondrocytes were used for the in vitro experiments. Destabilization of the medial meniscus (DMM) surgery was performed to establish an OA model in mice and IL‑1β was used to induce an OA model in vitro. The results indicated that AZ may promote chondrocyte viability and the expression of extracellular matrix components. Furthermore, AZ reduced the occurrence of ferroptosis by promoting the expression of glutathione peroxidase 4, inhibiting cartilage destruction and osteophyte formation, and alleviating damage to articular cartilage caused by DMM surgery. Mechanistically, the activation of nuclear factor erythroid 2‑related factor 2 and heme oxygenase‑1 may be responsible for the anti‑ferroptosis effects of AZ on chondrocytes. These findings indicated that AZ may be considered a promising candidate for OA therapy.
Collapse
Affiliation(s)
- Xu Chen
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Changzhou, Jiangsu 213003, P.R. China
| | - Jie Chen
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Changzhou, Jiangsu 213003, P.R. China
| | - Chunbao Miao
- Jiangsu Key Laboratory of Advanced Catalytic Materials and Technology, Jiangsu Province Key Laboratory of Fine Petrochemical Engineering, School of Petrochemical Engineering, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Guangrong Yin
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Changzhou, Jiangsu 213003, P.R. China
| | - Zhuangzhuang Zhang
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Changzhou, Jiangsu 213003, P.R. China
| | - Rongbin Sun
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Changzhou, Jiangsu 213003, P.R. China
| | - Su Ni
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
239
|
Chu C, Wang X, Yang C, Chen F, Shi L, Xu W, Wang K, Liu B, Wang C, Sun D, Ding W. Neutrophil extracellular traps drive intestinal microvascular endothelial ferroptosis by impairing Fundc1-dependent mitophagy. Redox Biol 2023; 67:102906. [PMID: 37812880 PMCID: PMC10579540 DOI: 10.1016/j.redox.2023.102906] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/11/2023] Open
Abstract
Microvascular endothelial damage caused by intestinal ischemia‒reperfusion (II/R) is a primary catalyst for microcirculation dysfunction and enterogenous infection. Previous studies have mainly focused on how neutrophil extracellular traps (NETs) and ferroptosis cause intestinal epithelial injury, and little attention has been given to how NETs, mainly from circulatory neutrophils, affect intestinal endothelial cells during II/R. This study aimed to unravel the mechanisms through which NETs cause intestinal microvascular dysfunction. We first detected heightened local NET infiltration around the intestinal microvasculature, accompanied by increased endothelial cell ferroptosis, resulting in microcirculation dysfunction in both human and animal II/R models. However, the administration of the ferroptosis inhibitor ferrostatin-1 or the inhibition of NETs via neutrophil-specific peptidylarginine deiminase 4 (Pad4) deficiency led to positive outcomes, with reduced intestinal endothelial ferroptosis and microvascular function recovery. Moreover, RNA-seq analysis revealed a significant enrichment of mitophagy- and ferroptosis-related signaling pathways in HUVECs incubated with NETs. Mechanistically, elevated NET formation induced Fundc1 phosphorylation at Tyr18 in intestinal endothelial cells, which led to mitophagy inhibition, mitochondrial quality control imbalance, and excessive mitochondrial ROS generation and lipid peroxidation, resulting in endothelial ferroptosis and microvascular dysfunction. Nevertheless, using the mitophagy activator urolithin A or AAV-Fundc1 transfection could reverse this process and ameliorate microvascular damage. We first demonstrate that increased NETosis could result in intestinal microcirculatory dysfunction and conclude that suppressed NET formation can mitigate intestinal endothelial ferroptosis by improving Fundc1-dependent mitophagy. Targeting NETs could be a promising approach for treating II/R-induced intestinal microcirculatory dysfunction.
Collapse
Affiliation(s)
- Chengnan Chu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xinyu Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chao Yang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Fang Chen
- School of Medicine, Southeast University, Nanjing, 210002, Jiangsu Province, China
| | - Lin Shi
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Weiqi Xu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Kai Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Baochen Liu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chenyang Wang
- Key Laboratory of Intestinal Injury, Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, PR China
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Weiwei Ding
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
240
|
Wang L, Liu Y, Li S, Zha Z, Chen Y, Wang Q, Zhou S, Huang X, Xu M. Capsaicin alleviates doxorubicin-induced acute myocardial injury by regulating iron homeostasis and PI3K-Akt signaling pathway. Aging (Albany NY) 2023; 15:11845-11859. [PMID: 37916995 PMCID: PMC10683596 DOI: 10.18632/aging.205138] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Capsaicin (CAP), a frequently occurring alkaloid component found in spicy peppers, has demonstrated therapeutic potential against tumors, metabolic disease, and cardiovascular disorders. Doxorubicin (DOX), a widely used anthracycline drug in chemotherapy, is notorious for its cardiotoxicity. This study aimed to investigate the potential of CAP in mitigating DOX toxicity in mouse hearts and H9C2 cells, as well as to explore the underlying mechanisms. METHODS In our study, we conducted experiments on both mice and H9C2 cells. The mice were divided into four groups and treated with different substances: normal saline, CAP, DOX and CAP+DOX. We evaluated the induction of ferroptosis by DOX and the remission of ferroptosis by CAP using various methods, including echocardiography, Hematoxylin and Eosin (H&E) staining, Masson's trichrome staining, and determination of ferroptosis metabolites, genes and proteins. Additionally, we employed RNA-seq to identify the inhibitory effect of CAP on DOX-induced myocardial apoptosis, which was further confirmed through western blotting. Similar approaches were applied to H9C2 cells, yielding reliable results. RESULTS Our study demonstrated that treatment with CAP improved the survival rate of DOX-treated mice and reduced myocardial injury. Mechanistically, CAP downregulated transferrin (Trf) and upregulated solute carrier family 40 member 1 (SLC40A1), which helped maintain iron levels in the cells and prevent ferroptosis. Furthermore, CAP inhibited DOX-induced apoptosis by modulating the phosphoinositide 3-kinase (PI3K)- protein kinase B (Akt) signaling pathway. Specifically, CAP activated the PI3K-Akt pathway and regulated downstream BCL2 and BAX to mitigate DOX-induced apoptosis. Therefore, our results suggest that CAP effectively alleviates acute myocardial injury induced by DOX. CONCLUSION Our findings demonstrate that CAP has the potential to alleviate DOX-induced ferroptosis by regulating iron homeostasis. Additionally, it can inhibit DOX-induced apoptosis by activating PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Longbin Wang
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ying Liu
- Department of Cardiology, Sixth Medical Center, PLA General Hospital, Beijing, China
| | - Si Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhijian Zha
- Chinese Internal Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yu Chen
- School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Shujing Zhou
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary, China
| | - Xufeng Huang
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary, China
| | - Ming Xu
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
241
|
Gu JJ, Du TJ, Zhang LN, Zhou J, Gu X, Zhu Y. Identification of Ferroptosis-Related Genes in Heart Failure Induced by Transverse Aortic Constriction. J Inflamm Res 2023; 16:4899-4912. [PMID: 37927963 PMCID: PMC10625389 DOI: 10.2147/jir.s433387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023] Open
Abstract
Background Heart failure (HF) is a common clinical syndrome due to ventricular dysfunction and is a major cause of mortality worldwide. Ferroptosis, marked by excessive iron-dependent lipid peroxidation, is closely related to HF. Therefore, the purpose of this study is to explore and validate ferroptosis-related markers in HF by bioinformatics analysis and animal experiments validation. Materials and Methods The gene expression profiles (GSE36074) of murine transverse aortic constriction (TAC) were obtained from the Gene Expression Omnibus (GEO); From the FerrDb database, ferroptosis-related genes (FRGs) were identified. Using GEO2R, differential expressed genes (DEGs) were screened. An overlapping analysis was conducted among DEGs and FRGs to identify ferroptosis-related DEGs (FRDEGs). We then performed clustering, functional enrichment analysis, and protein-protein interaction (PPI) analyses. In addition, the key FRDEGs were extracted by cytoHubba plugin and the networks of transcription factors (TFs)-key FRDEGs and microRNA-key FRDEGs were constructed. Lastly, the key FRDEGs were carried by quantitative reverse transcription PCR (RT-qPCR) and immunohistochemistry (IHC). Results Fifty-nine FRGs showing significantly different expression were identified from a total of 1918 DEGs in mice heart by transverse aortic constriction. GO and KEGG functional enrichment analysis revealed that these 59 ferroptosis-related DEGs mostly associated with positive regulation of apoptotic process, FoxO signaling pathway, VEGF signaling pathway, Apoptosis, Ferroptosis. Five key FRDEGs (Mapk14, Hif1a, Ddit3, Tlr4 and Ptgs2) were identified using PPI networks; Based on TFs-key FRDEGs networks, we found that Mapk14, Hif1a, Tlr4 and Ptgs2 were regulated by 3, 4, 5, and 29 TFs, respectively; however, Ddit3 was not regulated by any TF; By analyzing the miRNA-key FRDEGs networks, we found that 39, 74, 11, 28, and 18 miRNAs targets regulate the expression of Mapk14, Hif1a, Ddit3, Tlr4 and Ptgs2, respectively. Lastly, five key FRDEGs were validated at the mRNA and protein levels by RT-qPCR and IHC, which were in line with our bioinformatics analysis. Conclusion Our findings reveal that Mapk14, Hif1a, Ddit3, Tlr4 and Ptgs2 may be involved in the development of HF through regulating ferroptosis and as potential targets for HF.
Collapse
Affiliation(s)
- Jian Jun Gu
- Department of Cardiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Department of Cardiology, Northern Jiangsu People’s Hospital, Yangzhou, Jiangsu, People’s Republic of China
| | - Tian Jian Du
- Department of Cardiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Department of Cardiology, Northern Jiangsu People’s Hospital, Yangzhou, Jiangsu, People’s Republic of China
| | - Li Na Zhang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Jing Zhou
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Xiang Gu
- Department of Cardiology, Northern Jiangsu People’s Hospital, Yangzhou, Jiangsu, People’s Republic of China
| | - Ye Zhu
- Department of Cardiology, Northern Jiangsu People’s Hospital, Yangzhou, Jiangsu, People’s Republic of China
| |
Collapse
|
242
|
Cao D, Zheng J, Li Z, Yu Y, Chen Z, Wang Q. ACSL4 inhibition prevents macrophage ferroptosis and alleviates fibrosis in bleomycin-induced systemic sclerosis model. Arthritis Res Ther 2023; 25:212. [PMID: 37884942 PMCID: PMC10601156 DOI: 10.1186/s13075-023-03190-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Systemic sclerosis (SSc), with unclear pathophysiology, is a paradigmatic rheumatic disease of immunity dysfunction-driven multi-organ inflammation and ultimate fibrosis. Pathogenesis breakthroughs are urgently needed for available treatments halting its unremitting stiffness. This study aims to investigate whether ferroptosis can regulate the progressive SSc fibrosis. METHODS In vivo, bleomycin (BLM)-induced mice model was subjected to ferroptosis detection using western blotting, malondialdehyde (MDA), and glutathione (GSH) assays. Pharmacological inhibitor of the acyl-CoA synthetase long-chain family member 4 (ACSL4) was utilized to explore its potential therapeutic effects for fibrosis, from histological, biochemical, and molecular analyses. In vitro, bone marrow-derived macrophages (BMDM) were activated into inflammatory phenotype and then the relationship was evaluated between activation level and ferroptosis sensitivity in lipopolysaccharide (LPS) incubation with gradient concentrations. The potential calpain/ACSL4 axis was analyzed after calpain knockdown or over-expression in Raw264.7. RESULTS Both skin and lung tissue ferroptosis were present in SSc mice with enhanced ACSL4 expression, while ACSL4 inhibition effectively halted fibrosis progressing and provides protection from inflammatory milieu. Meanwhile, a positive regulation relationship between LPS-induced macrophage activity and ferroptosis sensitivity can be observed. After calpain knockdown, both inflammatory macrophage ferroptosis sensitivity and ACSL4 expression decreased, while its over-expression renders ACSL4-envoking condition. Also, calpain pharmacological inhibition reduced both ferroptosis and fibrosis aptitude in mice. CONCLUSIONS ACSL4 induces inflammatory macrophage ferroptosis to aggravate fibrosis progressing. ACSL4 and its upregulators of calpains may be potential therapeutic targets for BLM model of SSc.
Collapse
Affiliation(s)
- Dianyu Cao
- Department of Dermatology, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, 200032, P.R. China
| | - Jina Zheng
- Department of Dermatology, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, 200032, P.R. China
| | - Zheng Li
- Laboratory Animal Division, Institute of Clinical Science, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Yong Yu
- Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Zengrui Chen
- Department of Intensive Care Medicine, Yuhuan People's Hospital, No. 18 Changle Road, Yucheng Street, Yuhuan City, Zhejiang, 317600, P.R. China.
| | - Qiang Wang
- Department of Dermatology, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, 200032, P.R. China.
| |
Collapse
|
243
|
Wang X, Shen T, Lian J, Deng K, Qu C, Li E, Li G, Ren Y, Wang Z, Jiang Z, Sun X, Li X. Resveratrol reduces ROS-induced ferroptosis by activating SIRT3 and compensating the GSH/GPX4 pathway. Mol Med 2023; 29:137. [PMID: 37858064 PMCID: PMC10588250 DOI: 10.1186/s10020-023-00730-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 09/17/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Intestinal ischemia-reperfusion injury occurs in acute intestinal obstruction, intussusception, acute mesenteric artery embolism, and other diseases and can lead to local intestinal necrosis, distant organ involvement, or systemic reactions, with high morbidity and mortality. Ferroptosis plays a crucial role in intestinal ischemia-reperfusion injury, and inhibition of ferroptosis may provide new approaches for treating the disease. SIRT3 protects cells from oxidative stress and may be involved in the process of ferroptosis. We hypothesized that resveratrol, an agonist of SIRT3, could ameliorate intestinal ischemia-reperfusion injury by compensating the GSH/GPX4 pathway. METHODS Intestinal ischemia-reperfusion (I/R) and Caco-2 hypoxia-reoxygenation models were established. Transmission electron microscopy was used to assess mitochondrial function; the Chiu's score was used to evaluate the degree of intestinal mucosal injury based on HE staining; and Western blot was used to detect the SIRT3/FoxO3a pathway, tight junction proteins and ferroptosis-related protein expression. Sirt3-/- C57, shSIRT3-Caco-2 cells and siFoxO3a-Caco-2 cells were established. C11-BODIPY was used to detect lipid peroxide in cells; FD4 and IFABP were used to detect intestinal permeability; MitoSOX was used to detect ROS levels; and MitoTracker and immunofluorescence colocalization were used to detect SIRT3 levels. RESULTS In the intestinal I/R model, I/R injury occurs mainly during the reperfusion period and leads to ferroptosis through the GSH/GPX4 pathway. Resveratrol could reduce ferroptosis and ameliorate I/R injury by activating SIRT3. In Sirt3-/- mice, more intestinal mucosal cells underwent ferroptosis, I/R injury was more severe, and resveratrol lost the ability to ameliorate I/R injury. In addition, hypoxia-reoxygenation increased RSL3-induced ferroptosis sensitivity in Caco-2 cells in vitro. In the presence of shSIRT3 or RSL3 alone, resveratrol could ameliorate Caco-2 ferroptosis, but not RSL3-induced shSIRT3-Caco-2 ferroptosis. Furthermore, resveratrol might activate the SIRT3/FoxO3a pathway, increase the expression of SOD2 and catalase, and inhibit ROS generation, thus reducing lipid peroxidation and ferroptosis. CONCLUSION To date, this is the first study to show that resveratrol ameliorates intestinal ischemia-reperfusion injury by activating SIRT3 and reducing ferroptosis. Resveratrol can reduce intestinal ischemia-reperfusion injury by activating the SIRT3/FoxO3a pathway, increasing the expression of SOD2 and catalase, reducing ROS and LPO production, compensating for the GSH/GPX4 pathway and inhibiting ferroptosis. Resveratrol increases the expression of SOD2 and catalase, reduces the production of ROS and LPO, compensates for the GSH/GPX4 pathway and inhibits ferroptosis by activating the SIRT3/FoxO3a pathway.
Collapse
Affiliation(s)
- Xingjie Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Tianli Shen
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jie Lian
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Kai Deng
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Chao Qu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Enmeng Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Gan Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yiwei Ren
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zijun Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zhengdong Jiang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xuejun Sun
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
244
|
Feng S, Tang D, Wang Y, Li X, Bao H, Tang C, Dong X, Li X, Yang Q, Yan Y, Yin Z, Shang T, Zheng K, Huang X, Wei Z, Wang K, Qi S. The mechanism of ferroptosis and its related diseases. MOLECULAR BIOMEDICINE 2023; 4:33. [PMID: 37840106 PMCID: PMC10577123 DOI: 10.1186/s43556-023-00142-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023] Open
Abstract
Ferroptosis, a regulated form of cellular death characterized by the iron-mediated accumulation of lipid peroxides, provides a novel avenue for delving into the intersection of cellular metabolism, oxidative stress, and disease pathology. We have witnessed a mounting fascination with ferroptosis, attributed to its pivotal roles across diverse physiological and pathological conditions including developmental processes, metabolic dynamics, oncogenic pathways, neurodegenerative cascades, and traumatic tissue injuries. By unraveling the intricate underpinnings of the molecular machinery, pivotal contributors, intricate signaling conduits, and regulatory networks governing ferroptosis, researchers aim to bridge the gap between the intricacies of this unique mode of cellular death and its multifaceted implications for health and disease. In light of the rapidly advancing landscape of ferroptosis research, we present a comprehensive review aiming at the extensive implications of ferroptosis in the origins and progress of human diseases. This review concludes with a careful analysis of potential treatment approaches carefully designed to either inhibit or promote ferroptosis. Additionally, we have succinctly summarized the potential therapeutic targets and compounds that hold promise in targeting ferroptosis within various diseases. This pivotal facet underscores the burgeoning possibilities for manipulating ferroptosis as a therapeutic strategy. In summary, this review enriched the insights of both investigators and practitioners, while fostering an elevated comprehension of ferroptosis and its latent translational utilities. By revealing the basic processes and investigating treatment possibilities, this review provides a crucial resource for scientists and medical practitioners, aiding in a deep understanding of ferroptosis and its effects in various disease situations.
Collapse
Affiliation(s)
- Shijian Feng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dan Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yichang Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiang Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Bao
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengbing Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiuju Dong
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinna Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinxue Yang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun Yan
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhijie Yin
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tiantian Shang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Kaixuan Zheng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaofang Huang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zuheng Wei
- Chengdu Jinjiang Jiaxiang Foreign Languages High School, Chengdu, People's Republic of China
| | - Kunjie Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Shiqian Qi
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
245
|
Jin ZL, Gao WY, Guo F, Liao SJ, Hu MZ, Yu T, Yu SZ, Shi Q. Ring Finger Protein 146-mediated Long-chain Fatty-acid-Coenzyme a Ligase 4 Ubiquitination Regulates Ferroptosis-induced Neuronal Damage in Ischemic Stroke. Neuroscience 2023; 529:148-161. [PMID: 37591333 DOI: 10.1016/j.neuroscience.2023.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
Ischemic stroke (IS) is one of the leading causes of disability and death worldwide. Long-chain fatty-acid-coenzyme A ligase 4 (ACSL4) is a critical isozyme for ferroptosis that participates in the progression of IS. RING finger protein 146 (RNF146) is an E3 ligase predicted to interact with ACSL4 and regulated by activating transcription factor 3 (ATF3). The molecular mechanism of the RNF146/ACSL4 axis in IS is still unclear. Oxygen-glucose deprivation/reperfusion (OGD/R) treatment was used as the in vitro model, and middle cerebral artery occlusion (MCAO) mice were established for the in vivo model for IS. The protein level of ACSL4 was monitored by Western blot during ischemic injury. RNF146 was overexpressed in vitro and in vivo. The interaction of RNF146 and ACSL4 was determined by co-immunoprecipitation (Co-IP) assay. Chromatin immunoprecipitation (ChIP) assay and luciferase assay were utilized to determine the regulation of ATF3 on RNF146. Ferroptosis was evaluated by the levels of lactate dehydrogenase (LDH), malondialdehyde (MDA), Fe2+, and protein levels of related genes including ACSL4, SLC7A11, and GPX4. ACSL4 was downregulated upon OGD treatment and then increased by re-oxygenation. RNF146 was responsible for the ubiquitination and degradation of ACSL4 protein. RNF146 overexpression could prevent the stimulation of OGD/R-induced LDH, MDA, and Fe2+ levels and ferroptosis-related gene expression. ATF3 could activate the transcription and expression of RNF146, leading to the inhibition of OGD/R-induced neuron ferroptosis. The ATF3-mediated RNF146 could alleviate neuronal damage in IS by regulating ACSL4 ubiquitination and ferroptosis, providing a novel theoretical basis for exploring therapeutic targets and strategies.
Collapse
Affiliation(s)
- Zheng-Long Jin
- Department of Neurology, Affiliated Jiangmen Traditional Chinese Medicine Hospital of Ji'nan University, Jiangmen 529000, Guangdong Province, PR China
| | - Wen-Ying Gao
- Department of TCM Pediatrics, Jiangmen Maternal and Child Health Hospital, Jiangmen 529030, Guangdong Province, PR China
| | - Fu Guo
- Department of Neurology, Affiliated Jiangmen Traditional Chinese Medicine Hospital of Ji'nan University, Jiangmen 529000, Guangdong Province, PR China
| | - Shao-Jun Liao
- Department of Spine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, Guangdong Province, PR China
| | - Ming-Zhe Hu
- Department of Neurology, The Affiliated Hospital of Shandong University of TCM, Jinan 250000, Shandong Province, PR China
| | - Tao Yu
- Department of Neurology, Affiliated Jiangmen Traditional Chinese Medicine Hospital of Ji'nan University, Jiangmen 529000, Guangdong Province, PR China
| | - Shang-Zhen Yu
- Department of Neurology, Affiliated Jiangmen Traditional Chinese Medicine Hospital of Ji'nan University, Jiangmen 529000, Guangdong Province, PR China
| | - Qing Shi
- Department of Neurology, Affiliated Jiangmen Traditional Chinese Medicine Hospital of Ji'nan University, Jiangmen 529000, Guangdong Province, PR China.
| |
Collapse
|
246
|
Yu T, Sun S. Role and mechanism of ferroptosis in acute lung injury. Cell Cycle 2023; 22:2119-2129. [PMID: 37946318 PMCID: PMC10732650 DOI: 10.1080/15384101.2023.2278328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/28/2023] [Indexed: 11/12/2023] Open
Abstract
Ferroptosis is a new non-apoptotic cell death caused by the accumulation of dysregulated metabolism of ferric iron, amino acids or lipid peroxidation. Increasing studies suggest that ferroptosis is involved in the acute lung injury (ALI). This article aims to review the role of ferroptosis in ALI. ALI is a common respiratory disease and presents a high mortality rate. Inhibiting cell ferroptosis of lung improves the ALI. In addition, several signaling pathways are related to ferroptosis in ALI, involving in iron homeostasis, lipid peroxidation, and amino acid metabolism. Moreover, there are various key factors to regulate the occurrence of ferroptosis in ALI, such as ACSL4, NRF2, and P53. The ACSL4 promotes the ferroptosis, while the NRF2 alleviates the ferroptosis in ALI. The main effect of P53 is to promote ferroptosis. Accordingly, ferroptosis is involved in ALI and may be an important therapeutic target for ALI.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
- Pediatrics Class 1, Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
247
|
Liu ZL, Huang YP, Wang X, He YX, Li J, Li B. The role of ferroptosis in chronic intermittent hypoxia-induced cognitive impairment. Sleep Breath 2023; 27:1725-1732. [PMID: 36607542 DOI: 10.1007/s11325-022-02760-6] [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: 08/25/2022] [Revised: 11/14/2022] [Accepted: 12/01/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Obstructive sleep apnea (OSA) is a sleep disorder that may lead to cognitive impairment. The primary pathophysiological feature of OSA is chronic intermittent hypoxia (CIH), but the underlying mechanisms of CIH are not known. There have been few studies on the role of ferroptosis, a novel form of programmed cell death, during CIH-induced cognitive impairment. Therefore, this paper examined ferroptosis' effect on CIH-mediated cognitive impairment. METHODS The study randomized twenty-four Sprague-Dawley (SD) male rats to control or CIH group. CIH rats were subjected to intermittent hypoxia for 4 weeks. Rat learning and memory were analyzed by the Morris water maze (MWM) test. Alterations of hippocampal neuronal ultrastructure were observed by transmission electron microscopy (TEM). Malondialdehyde (MDA) and ferrous iron (Fe2+) levels and superoxide dismutase (SOD) and reduced glutathione (GSH) contents were determined. Ferroptosis-associated protein levels were examined by Western blotting. RESULTS The MWM test indicated that rats in the CIH group exhibited neurocognitive impairment. TEM showed that CIH induced mitochondrial damage. Significant increases in Fe2+ and MDA levels were observed in the CIH group, and GSH and SOD levels were decreased. Expression of Acyl-CoA synthetase long-chain family member 4 (ACSL4) increased, and glutathione peroxidase 4 (GPX4) protein levels were decreased, suggesting that ferroptosis was induced in CIH model rats. The NF-E2-related factor 2 (Nrf2) protein level in the CIH group was decreased. CONCLUSION Ferroptosis had an essential effect on CIH-mediated cognitive impairment, and it may occur via Nrf2 dysregulation. These findings lay a solid foundation for the subsequent study of OSA-associated cognitive impairment offering potential evidence for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Zhi-Li Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yin-Pei Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xin Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yu-Xin He
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Juan Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Bing Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
248
|
Gao X, Hu W, Qian D, Bai X, He H, Li L, Sun S. The Mechanisms of Ferroptosis Under Hypoxia. Cell Mol Neurobiol 2023; 43:3329-3341. [PMID: 37458878 PMCID: PMC10477166 DOI: 10.1007/s10571-023-01388-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/06/2023] [Indexed: 09/05/2023]
Abstract
Ferroptosis is a new form of programmed cell death, which is characterized by the iron-dependent accumulation of lipid peroxidation and increase of ROS, resulting in oxidative stress and cell death. Iron, lipid, and multiple signaling pathways precisely control the occurrence and implementation of ferroptosis. The pathways mainly include Nrf2/HO-1 signaling pathway, p62/Keap1/Nrf2 signaling pathway. Activating p62/Keap1/Nrf2 signaling pathway inhibits ferroptosis. Nrf2/HO-1 signaling pathway promotes ferroptosis. Furthermore, some factors also participate in the occurrence of ferroptosis under hypoxia, such as HIF-1, NCOA4, DMT1. Meanwhile, ferroptosis is related with hypoxia-related diseases, such as MIRI, cancers, and AKI. Accordingly, ferroptosis appears to be a therapeutic target for hypoxia-related diseases.
Collapse
Affiliation(s)
- Xin Gao
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, 650032, China
- 2020 Clinical Medicine Class 6, Kunming Medical University, Kunming, 650500, China
| | - Wei Hu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China
| | - Dianlun Qian
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| | - Xiangfeng Bai
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| | - Huilin He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, 650032, China
| | - Lin Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, 650032, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, 650032, China.
| |
Collapse
|
249
|
Zhao J, Li J, Wei D, Gao F, Yang X, Yue B, Xiong D, Liu M, Xu H, Hu C, Chen J. Liproxstatin-1 Alleviates Lung Transplantation-induced Cold Ischemia-Reperfusion Injury by Inhibiting Ferroptosis. Transplantation 2023; 107:2190-2202. [PMID: 37202851 DOI: 10.1097/tp.0000000000004638] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
BACKGROUND Primary graft dysfunction, which is directly related to cold ischemia-reperfusion (CI/R) injury, is a major obstacle in lung transplantation (LTx). Ferroptosis, a novel mode of cell death elicited by iron-dependent lipid peroxidation, has been implicated in ischemic events. This study aimed to investigate the role of ferroptosis in LTx-CI/R injury and the effectiveness of liproxstatin-1 (Lip-1), a ferroptosis inhibitor, in alleviating LTx-CI/R injury. METHODS LTx-CI/R-induced signal pathway alterations, tissue injury, cell death, inflammatory responses, and ferroptotic features were examined in human lung biopsies, the human bronchial epithelial (BEAS-2B) cells, and the mouse LTx-CI/R model (24-h CI/4-h R). The therapeutic efficacy of Lip-1 was explored and validated both in vitro and in vivo. RESULTS In human lung tissues, LTx-CI/R activated ferroptosis-related signaling pathway, increased the tissue iron content and lipid peroxidation accumulation, and altered key protein (GPX4, COX2, Nrf2, and SLC7A11) expression and mitochondrial morphology. In BEAS-2B cells, the hallmarks of ferroptosis were significantly evidenced at the setting of both CI and CI/R compared with the control, and the effect of adding Lip-1 only during CI was much better than that of only during reperfusion by Cell Counting Kit-8. Furthermore, Lip-1 administration during CI markedly relieved LTx-CI/R injury in mice, as indicated by significant improvement in lung pathological changes, pulmonary function, inflammation, and ferroptosis. CONCLUSIONS This study revealed the existence of ferroptosis in the pathophysiology of LTx-CI/R injury. Using Lip-1 to inhibit ferroptosis during CI could ameliorate LTx-CI/R injury, suggesting that Lip-1 administration might be proposed as a new strategy for organ preservation.
Collapse
Affiliation(s)
- Jin Zhao
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jiawei Li
- Department of Intensive Care Medicine, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Dong Wei
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Fei Gao
- Department of Emergency, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Xiucheng Yang
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Bingqing Yue
- Department of Lung Transplantation, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Dian Xiong
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Mingzhao Liu
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Hongyang Xu
- Department of Intensive Care Medicine, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Chunxiao Hu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jingyu Chen
- Wuxi Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
- Department of Lung Transplantation, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
250
|
Li L, Ye Z, Xia Y, Li B, Chen L, Yan X, Yuan T, Song B, Yu W, Rao T, Lin F, Zhou X, Cheng F. YAP/ACSL4 Pathway-Mediated Ferroptosis Promotes Renal Fibrosis in the Presence of Kidney Stones. Biomedicines 2023; 11:2692. [PMID: 37893066 PMCID: PMC10603838 DOI: 10.3390/biomedicines11102692] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/20/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
The potential association between calcium oxalate stones and renal fibrosis has been extensively investigated; however, the underlying mechanisms remain unclear. Ferroptosis is a novel form of cell death characterized by iron-dependent lipid peroxidation and regulated by acyl coenzyme A synthase long-chain family member 4 (ACSL4). Yes-associated protein (YAP), a transcriptional co-activator in the Hippo pathway, promotes ferroptosis by modulating ACSL4 expression. Nevertheless, the involvement of YAP-ACSL4 axis-mediated ferroptosis in calcium oxalate crystal deposition-induced renal fibrosis and its molecular mechanisms have not been elucidated. In this study, we investigated ACSL4 expression and ferroptosis activation in the kidney tissues of patients with calcium oxalate stones and in mice using single-cell sequencing, transcriptome RNA sequencing, immunohistochemical analysis, and Western blot analysis. In vivo and in vitro experiments demonstrated that inhibiting ferroptosis or ACSL4 mitigated calcium oxalate crystal-induced renal fibrosis. Furthermore, YAP expression was elevated in the kidney tissues of patients with calcium oxalate stones and in calcium oxalate crystal-stimulated human renal tubular epithelial cell lines. Mechanistically, in calcium oxalate crystal-stimulated human renal tubular epithelial cell lines, activated YAP translocated to the nucleus and enhanced ACSL4 expression, consequently inducing cellular ferroptosis. Moreover, YAP silencing suppressed ferroptosis by downregulating ACSL4 expression, thereby attenuating calcium oxalate crystal-induced renal fibrosis. Conclusively, our findings suggest that YAP-ACSL4-mediated ferroptosis represents an important mechanism underlying the induction of renal fibrosis by calcium oxalate crystal deposition. Targeting the YAP-ACSL4 axis and ferroptosis may therefore hold promise as a potential therapeutic approach for preventing renal fibrosis in patients with kidney stones.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (L.L.); (Z.Y.)
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (L.L.); (Z.Y.)
| |
Collapse
|