1
|
Ju Y, Ma C, Huang L, Tao Y, Li T, Li H, Huycke MM, Yang Y, Wang X. Inactivation of glutathione S-transferase alpha 4 blocks Enterococcus faecalis-induced bystander effect by promoting macrophage ferroptosis. Gut Microbes 2025; 17:2451090. [PMID: 39819335 PMCID: PMC11740687 DOI: 10.1080/19490976.2025.2451090] [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: 05/14/2024] [Revised: 11/08/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025] Open
Abstract
Enterococcus faecalis-infected macrophages produce 4-hydroxynonenal (4-HNE) that mediates microbiota-induced bystander effect (MIBE) leading to colorectal cancer (CRC). Glutathione S-transferase alpha 4 (Gsta4), a specific detoxifying enzyme for 4-HNE, is overexpressed in human CRC and E. faecalis-induced murine CRC. However, the roles of Gsta4 in E. faecalis-induced colitis and CRC remain unclear. Herein, we demonstrate that Gsta4 is essential for MIBE by protecting macrophages from E. faecalis-induced ferroptosis. E. faecalis OG1RFSS was used to induce colitis in Gsta4-/- and Il10-/-/Gsta4-/- mice by orogastric gavage. Ferroptosis was assessed in Gsta4-deficient murine macrophages. We found that, unlike Il10-/- mice, Gsta4-/- and Il10-/-/Gsta4-/- mice colonized with E. faecalis failed to develop colitis or CRC. Immunofluorescent staining showed a reduction of macrophages in the lamina propria of E. faecalis-colonized Il10-/-/Gsta4-/- mice, as well as decreased Gpx4 expression, indicating the occurrence of ferroptosis. Ferroptosis was further confirmed in Gsta4-deficient murine macrophages infected with E. faecalis. Moreover, Gsta4 inactivation induced the upregulation of Hmox1 and phosphorylated c-Jun while blocked Nos2 expression, leading to the accumulation of intracellular ferrous iron, lipid peroxidation and, eventually, ferroptosis. Finally, Mapk8, as a ferroptosis driver, was remarkably elevated in E. faecalis-infected Gsta4-deficient macrophages. These results suggest that Gsta4 inactivation blocks MIBE by eliminating macrophages, thereby attenuates E. faecalis-induced colitis and CRC.
Collapse
Affiliation(s)
- Yuanyuan Ju
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Genetics and Reproductive Medicine, Nantong, Jiangsu, China
| | - Chunhua Ma
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Genetics and Reproductive Medicine, Nantong, Jiangsu, China
| | - Lin Huang
- Department of Gastroenterology, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yumei Tao
- Department of Pathology, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
| | - Tianqi Li
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Genetics and Reproductive Medicine, Nantong, Jiangsu, China
| | - Haibo Li
- Department of Clinical Laboratory, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
| | - Mark M. Huycke
- Stephenson Cancer Center, Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yonghong Yang
- Department of Nephrology, Rheumatology, and Immunology, Nantong Children’s Hospital, Nantong, Jiangsu, China
- Department of Pediatrics, Nantong Maternity and Child Healthcare Hospital, Nantong, Jiangsu, China
| | - Xingmin Wang
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Genetics and Reproductive Medicine, Nantong, Jiangsu, China
- Stephenson Cancer Center, Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
2
|
Xu Z, Zhang M, Zhang X, Han H, Ye W, Chen Z, Lv Z, Liu Y, Liu Z, Gong J, Zhu B, Zhou S, Zhu R, Tao C, Zhang G, Yan X. Dihydromyricetin protects against cisplatin-induced renal injury and mitochondria-mediated apoptosis via the EGFR/HSP27/STAT3 signaling pathway. Ren Fail 2025; 47:2490202. [PMID: 40230054 PMCID: PMC12001862 DOI: 10.1080/0886022x.2025.2490202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 03/06/2025] [Accepted: 03/22/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Cisplatin (CP) has been used as an effective chemotherapy drug for different types of cancers. Despite its therapeutic benefits, the clinical utility of CP is often hindered by adverse effects, notably acute kidney injury (AKI), which restricts its widespread application. Dihydromyricetin (DHM) is a flavonoid acquired from Ampelopsis grossedentata, exhibiting a range of pharmacological activities. The major objective of this research was to examine the possible molecular mechanism involved in CP-induced AKI and the protective function of DHM. METHODS In this study, the protective function of DHM against CP-induced AKI was assessed in both mice and HK-2 cells. Kidney dysfunction parameters and renal morphology were evaluated to ascertain the extent of protection. Additionally, proteomics techniques were employed to investigate the protective effect of DHM and elucidate the underlying molecular mechanisms involved in mitigating CP-induced AKI. In addition, protein levels of epidermal growth factor receptor (EGFR), p-EGFR, heat shock protein 27 (HSP27), p-HSP27, STAT3, and p-STAT3 in renal tissues were investigated. Furthermore, an EGFR-blocking agent (gefitinib) or si-RNA of HSP27 was used to study the effects of inhibiting EGFR or HSP27 on CP-induced renal injury. RESULTS DHM decreased blood urea nitrogen (BUN) and creatinine in serum, alleviated renal morphological injury and downregulated the expression of CP-induced kidney injury molecule-1 and neutrophil gelatinase-related lipocalin. Proteomic data revealed HSP27 as a potential therapeutic target for AKI. DHM treatment resulted in the downregulation of EGFR, HSP27, and STAT3 phosphorylation, ultimately mitigating CP-induced AKI. In addition, the inhibition of EGFR or HSP27 reduced mitochondria-mediated apoptosis and CP-induced cell damage in HK-2 cells. CONCLUSIONS DHM effectively inhibited CP-induced oxidative stress, inflammation, and mitochondria-mediated apoptosis through the EGFR/HSP27/STAT3 pathway.
Collapse
Affiliation(s)
- Zheming Xu
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Minjing Zhang
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Xue Zhang
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Huirong Han
- School of Anesthesiology, Shandong Second Medical University, Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, Weifang, China
| | - Weifeng Ye
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Zhenjie Chen
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Zhisu Lv
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Yang Liu
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Zhengye Liu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianguang Gong
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Bin Zhu
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Suhan Zhou
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Runzhi Zhu
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Chang Tao
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Gensheng Zhang
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Xiang Yan
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| |
Collapse
|
3
|
Cui QF, Liu C, Dong XM, Liu ZQ. Exploring the biological functions and disease implications of OSGINs: A journey from discovery to clinical relevance. Biochem Pharmacol 2025; 237:116921. [PMID: 40199404 DOI: 10.1016/j.bcp.2025.116921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
Oxidative stress-induced growth inhibitors (OSGINs) represent a new category of proteins that respond to oxidative stress and modulate redox balance. Growing evidence indicates that OSGINs have extensive physiological and pathological functions by regulating essential cellular processes, including proliferation, autophagy, apoptosis, and ferroptosis, thus influencing the progression of various diseases such as cancer, atherosclerosis, and pulmonary fibrosis. Moreover, research indicates that some contaminants, biomaterials, active compounds, and drugs can induce the expression of OSGINs, thereby exerting toxicity or therapeutic effects on the organism. These many functions make OSGINs attractive targets. However, a thorough analysis of the topic is still lacking. This paper presents a systematic review of current OSGINs research, with an emphasis on their molecular functions, regulatory mechanisms, disease roles, and environmental stressors. Furthermore, using virtual screening tools, we identified a series of active molecules with potential inhibitory effects on OSGINs, providing valuable references for further drug development. Our review presents novel insights and guidance for the ongoing investigation of the biological significance and potential clinical applications of OSGINs.
Collapse
Affiliation(s)
- Qian-Fei Cui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Chong Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xue-Man Dong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhao-Qian Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
4
|
Tian Z, Wu Y, Yi B, Li L, Liu Y, Zhang H, Li A. ESCRT III-mediated lysosomal repair improve renal tubular cell injury in cisplatin-induced AKI. Autophagy 2025:1-18. [PMID: 40152606 DOI: 10.1080/15548627.2025.2483598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
The chemotherapeutic agent cisplatin is widely utilized for the treatment of various solid tumors. However, its clinical utility is limited by nephrotoxicity. Although numerous studies have demonstrated the potential of enhancing macroautophagy/autophagy in alleviating cisplatin-induced acute kidney injury (AKI), the dynamics of the autophagic process during renal tubular injury remain to be elucidated. In our investigation, we observed that cisplatin treatment leads to increased expression of LC3-II, GABARAPL1, SQSTM1/p62 and NBR1 in mouse renal tubular epithelial cells and BUMPT cells. Moreover, ultrastructurally, there is extensive accumulation of autophagic vacuoles in AKI mice. These findings imply that cisplatin-induced AKI results in impaired autophagic flow within renal tubular cells. Furthermore, LGALS3 (galectin 3) was found to be enriched in lysosomes after cisplatin treatment, revealing a close association between autophagy dysfunction and impaired lysosomal membrane integrity. Given the damaging contents of lysosomes, lysosomal membrane permeabilization must be rapidly resolved. Our findings showed that ESCRT III subunit CHMP4A-mediated lysosomal membrane repair significantly ameliorates autophagic defects and protects against lysosomal damage-induced cell death in a cisplatin-induced AKI model. In conclusion, our study indicates that ESCRT III-mediated lysosomal repair can relieve cisplatin-induced cell apoptosis and restore normal autophagy function in renal tubular epithelial cells. This mechanism plays a protective role against cisplatin-induced AKI.Abbreviations: AAV: adeno-associated virus; AKI: acute kidney injury; CQ: chloroquine; ESCRT: endosomal sorting complex required for transport; LMP: lysosomal membrane permeabilization; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PAS: periodic acid Schiff; PTECs: proximal renal tubule epithelial cells; TEM: transmission electron microscopy; TUNEL: terminal deoxynucleotidyl transferase dUTP nick end labeling.
Collapse
Affiliation(s)
- Zhangyu Tian
- Department of Nephrology, The Third Xiangya Hospital, The Critical Kidney Disease Research Center, Central South University, Changsha, Hunan, China
| | - Yiming Wu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Bin Yi
- Department of Nephrology, The Third Xiangya Hospital, The Critical Kidney Disease Research Center, Central South University, Changsha, Hunan, China
| | - Ling Li
- Department of Nephrology, The Third Xiangya Hospital, The Critical Kidney Disease Research Center, Central South University, Changsha, Hunan, China
| | - Yan Liu
- Department of Nephrology, The Third Xiangya Hospital, The Critical Kidney Disease Research Center, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, The Critical Kidney Disease Research Center, Central South University, Changsha, Hunan, China
| | - Aimei Li
- Department of Nephrology, The Third Xiangya Hospital, The Critical Kidney Disease Research Center, Central South University, Changsha, Hunan, China
| |
Collapse
|
5
|
Chu Y, Wei M, Cao Z, Chen L, Tan J, Bao W, Yang F, Zhang Y, Lin Y, Zhang Y, Li S, Lv C, Zhou W, Du H, Shen L, Huai C, Wang Z, Qin S. Integrative analysis based on CRISPR screen identifies apilimod as a potential therapeutic agent for cisplatin-induced acute kidney injury treatment. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-025-2874-8. [PMID: 40138089 DOI: 10.1007/s11427-025-2874-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025]
Abstract
Acute kidney injury (AKI), a life-threatening side effect of cisplatin therapy, significantly limits the drug's therapeutic potential. In this study, we conducted a genome-wide CRISPR/Cas9 knockout screen in human renal tubular epithelial cells, integrating the results with transcriptome analyses and the Connectivity Map (CMap) database. Apilimod and elacridar emerged as the top two candidates of mitigating cisplatin-induced nephrotoxicity, with apilimod demonstrating superior efficacy in drug matrix experiments. Apilimod reduced cisplatin-induced apoptosis, inflammation and reactive oxygen species (ROS) generation. Transcriptome analyses suggested that apilimod may protect against cisplatin-induced nephrotoxicity via modulating lipid metabolism. In vitro experiments revealed that apilimod significantly ameliorated cisplatin-induced lipotoxicity by enhancing lipid clearance and upregulating PGC1α-mediated fatty acid oxidation. Mechanism experiments showed that apilimod induces the nuclear translocation of TFEB through the inhibition of its target, PIKfyve, thereby enhancing PGC1α expression and ameliorating lipotoxicity. These protective effects of apilimod were simulated by siRNA-mediated PIKfyve knockdown and diminished by the PGC1α inhibitor SR-18292 and siRNA targeting TFEB, confirming the role of the PIKfyve/TFEB/PGC1α signaling axis in apilimod's renoprotective effects. In vivo, apilimod alleviated apoptosis, inflammation, and lipid accumulation in a cisplatin-induced AKI mouse model. Additionally, apilimod treatment did not compromise the antitumor effect of cisplatin in cancer cells or tumor-bearing mice. Overall, our study suggests that apilimod could be a promising therapeutic agent for the treatment of cisplatin-induced AKI and revealed its underlying molecular mechanism.
Collapse
Affiliation(s)
- Yunpeng Chu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Muyun Wei
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Baoshan Branch, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201900, China
| | - Zhongyu Cao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Luan Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Jie Tan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Wei Bao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Fan Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Yingtian Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Yunxiao Lin
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Yutong Zhang
- School of Life Sciences and Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shiyi Li
- School of Life Sciences and Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Cai Lv
- Department of Urology, Affiliated Haikou Hospital of Xiangya Medical School, Central South University, Haikou, 100062, China
| | - Wei Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Huihui Du
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Lu Shen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Cong Huai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China
| | - Zhenting Wang
- Department of Urology, Affiliated Haikou Hospital of Xiangya Medical School, Central South University, Haikou, 100062, China.
| | - Shengying Qin
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200230, China.
| |
Collapse
|
6
|
Zhang Y, Jing Y, He J, Dong R, Li T, Li F, Zheng X, Liu G, Jia R, Xu J, Wu F, Jia C, Song J, Zhang L, Zhou P, Wang H, Yao Z, Liu Q, Yu Y, Zhou J. Bile acid receptor FXR promotes intestinal epithelial ferroptosis and subsequent ILC3 dysfunction in neonatal necrotizing enterocolitis. Immunity 2025; 58:683-700.e10. [PMID: 40023163 DOI: 10.1016/j.immuni.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 11/14/2024] [Accepted: 02/04/2025] [Indexed: 03/04/2025]
Abstract
Necrotizing enterocolitis (NEC) is a common pediatric emergency primarily afflicting preterm infants, yet its mechanisms remain to be fully understood. Here, we report that plasma fibroblast growth factor (FGF)19, a target of farnesoid X receptor (FXR), was positively correlated with the clinical parameters of NEC. NEC patients and the NEC murine model displayed abundant FXR in intestinal epithelial cells (IECs), which was restricted by microbiota-derived short-chain fatty acids (SCFAs) under homeostasis. Genetic deficiency of FXR in IECs caused remission of NEC. Mechanistically, FXR facilitated ferroptosis of IECs via targeting acyl-coenzyme A synthetase long-chain family member 4 (Acsl4). Lipid peroxides released by ferroptotic IECs suppressed interleukin (IL)-22 secretion from type 3 innate lymphoid cells (ILC3s), thereby exacerbating NEC. Intestinal FXR antagonist, ACSL4 inhibitor, and ferroptosis inhibitor ameliorated murine NEC. Furthermore, the elevated lipid peroxides in NEC patients were positively correlated with FGF19 and disease parameters. These observations demonstrate the therapeutic value of targeting intestinal FXR and ferroptosis in NEC treatment.
Collapse
Affiliation(s)
- Yuxin Zhang
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China
| | - Yuchao Jing
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China; Department of Immunology, Basic Medical College, Changzhi 046000, China
| | - Juan He
- Provincial Key Laboratory of Research in Structure Birth Defect Disease and Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Rui Dong
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China
| | - Tongyang Li
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China
| | - Fang Li
- Department of Central Laboratory, Changzhi Medical College, Changzhi 046000, China
| | - Xiaoqing Zheng
- Laboratory of Immunity, Inflammation & Cancer, Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Gaoyu Liu
- Laboratory of Immunity, Inflammation & Cancer, Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ran Jia
- Department of Clinical Laboratory, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Jin Xu
- Department of Clinical Laboratory, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Fan Wu
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, Guangdong, China
| | - Chunhong Jia
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, Guangdong, China
| | - Jin Song
- Department of Pediatric Surgery Maternal and Child Health Care of Changzhi, Changzhi 046011, China
| | - Lijuan Zhang
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China
| | - Pan Zhou
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China
| | - Haitao Wang
- Department of Oncology, The Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Precision Medicine for Sex Hormones and Diseases, Tianjin 300211, China
| | - Zhi Yao
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China
| | - Qiang Liu
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China; Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| | - Ying Yu
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China; Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jie Zhou
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin 300070, China; Laboratory of Immunity, Inflammation & Cancer, Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
7
|
Zhang Y, Hu J, Zhang Y, Ci X. Amentoflavone protects against cisplatin-induced acute kidney injury by modulating Nrf2-mediated oxidative stress and ferroptosis and partially by activating Nrf2-dependent PANoptosis. Front Pharmacol 2025; 16:1508047. [PMID: 40110131 PMCID: PMC11919867 DOI: 10.3389/fphar.2025.1508047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Background Cisplatin is a widely used drug for the treatment of solid organ cancer, but its renal toxicity cannot be ignored. Amentoflavone (AME), a natural flavonoid compound, has remarkable pharmacological effects, including anti-inflammatory and antioxidative effects. The effect and mechanism of AME on cisplatin-induced acute kidney injury (CI-AKI) remain unclear. Methods We investigated the effect of AME on CI-AKI using the HK-2 cell line and C57BL/6 mice. Renal function, tissue damage, and molecular markers were assessed to explore the effects of AME on oxidative stress and cell death pathways. Results In vitro, AME significantly suppressed the cytotoxic effects of cisplatin on HK-2 cells. Furthermore, AME significantly inhibited cisplatin-induced ferroptosis and PANoptosis (apoptosis, pyroptosis and necroptosis). In mice with acute kidney injury induced by a single intraperitoneal injection of cisplatin, the daily administration of AME during AKI effectively improved renal function and alleviated renal tubular injury, characterized by the normalization of blood urea nitrogen (BUN) and serum creatinine (SCr) levels; it also inhibited cisplatin-induced renal ferroptosis and PANoptosis. AME is a natural antioxidant that activates the Nrf2 antioxidant pathway both in vivo and in vitro. In Nrf2 knockout mice and knockdown cells, the protective effect of AME against cisplatin-induced nephrotoxicity disappeared. However, after Nrf2 knockout, the effect of AME on ferroptosis completely disappeared, and that on PANoptosis partially disappeared. Conclusion Amentoflavone has a protective effect on cisplatin-induced acute kidney injury via a mechanism related to the Nrf2-dependent antioxidant pathway and the regulation of ferroptosis and PANoptosis.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Jianqiang Hu
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Yanmin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- Jilin Provincial Key Laboratory of Women's Reproductive Health, Changchun, Jilin, China
| |
Collapse
|
8
|
Althagafy HS, Hassanein EHM. Morin Mitigates 5-Fluorouracil-Induced Nephrotoxicity by Activating Nrf2/HO-1 and FXR, and Suppressing ERK/VCAM-1 and NF-κB Pathways. Int Immunopharmacol 2025; 148:114092. [PMID: 39823795 DOI: 10.1016/j.intimp.2025.114092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/06/2024] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
5-Fluorouracil (5-FU) is a DNA analog used in chemotherapy to treat various tumors. However, the clinical use of 5-FU is limited due to its severe adverse effects, particularly its nephrotoxicity. Morin (MRN) is a flavanol found in many different plants, including those in the Moraceae family, and has anti-inflammatory and antioxidant bioactivities. The protective effects of MRN against experimental 5-FU-induced kidney injury were investigated in this work. The rats were assigned to four groups in our study: control, MRN (50 mg/kg), 5-FU (30 mg/kg), and 5-FU + MRN. The administration of MRN caused a significant (P < 0.05) decrease in the serum urea and creatinine levels and a reduction in the histopathological changes induced by 5-FU, as shown by H&E, PAS, and Sirius red staining. IHC shows that MRN attenuates renal oxidative stress induced by 5-FU via co-activation of Nrf2, HO-1, and FXR. MRN protects against renal inflammation induced by 5-FU, as evidenced by decreased TNF-α and IL-6 levels in the rat kidney mediated by the downregulation of the ERK1/2 and VCAM-1 proteins and decreased NF-κB phosphorylation as shown by Western blotting. These findings support using MRN as a novel and promising treatment for 5-FU-induced nephrotoxicity.
Collapse
Affiliation(s)
- Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Saudi Arabia.
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| |
Collapse
|
9
|
Hu J, Zhang Y, Zhang Y, Shi N, Miu Y, Huang J, Miao M, Ci X. Bergenin inhibits ferritinophagy and ferroptosis in cisplatin-induced acute kidney injury by activating the p-GSK3β/Nrf2/PPARγ pathway. Int Immunopharmacol 2025; 147:114004. [PMID: 39793228 DOI: 10.1016/j.intimp.2024.114004] [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: 10/28/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
Ferroptosis plays a key role in cisplatin-induced acute kidney injury (AKI). Bergenin, which is extracted from Ardisiae Japonicae Herba and has long been used in folk tea and herbal tea drinks, is known to activate Nrf2 and has anti-inflammatory and antioxidant properties, however, its protective influence on CI-AKI has not been elucidated. We used models of cisplatin-induced nephrotoxicity in vitro and CI-AKI models in vivo. In vitro, we found that ferroptosis and ferritinophagy biomarkers were strongly regulated by bergenin treatment. Mechanistic experiments demonstrated that bergenin bound to and phosphorylated GSK3β, which inhibited its activity, to promote the nuclear translocation of Nrf2 and its subsequent binding to the PPARγ promoter sequence to activate PPARγ. However, the protective effects of bergenin on ferroptosis and ferritinophagy in cisplatin-exposed HK-2 cells were diminished when Nrf2 or PPARγ was inhibited. In vivo, bergenin effectively inhibited renal damage induced by cisplatin. Furthermore, bergenin attenuated ferritinophagy-mediated ferroptosis caused by cisplatin; these effects were abolished in Nrf2 knockout mice. Our findings revealed that bergenin effectively protected against ferritinophagy and ferroptosis in CI-AKI, which was largely dependent on the activation of the p-GSK3β/Nrf2/PPARγ pathway.
Collapse
Affiliation(s)
- Jianqiang Hu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Yan Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Yanmin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Ningmohan Shi
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Yufan Miu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Jing Huang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Mochi Miao
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China.
| |
Collapse
|
10
|
Dong H, Zhu L, Sun J, Chen Q, Liu P, Zhang W, Zeng H, Lin R, Yu Z, Lu J. IFNβ drives ferroptosis through elevating TRIM22 and promotes the cytotoxicity of RSL3. Front Immunol 2025; 16:1535554. [PMID: 39975542 PMCID: PMC11836015 DOI: 10.3389/fimmu.2025.1535554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
Background Cyclic GMP-AMP synthase (cGAS)-stimulator-of-interferon genes (STING) pathway is a cytosolic DNA sensor system. The production of this pathway, interferon-β (IFNβ), could suppress the growth of tumor cells, yet it is unclear whether ferroptosis is involved in IFNβ-induced cell death. Methods The effects of IFNβ on ferroptosis were analyzed in HT1080, 4T1, HCT116 and 786-O cells. HT1080 and 4T1 cells treated with IFNβ were subjected to RNA-Seq analysis. STAT1, STAT3, TRIM21, and TRIM22 were silenced by siRNAs to examine their effects on IFNβ-induced ferroptosis. The cGAS-STING signaling pathway-activated mice were used to evaluate the effects of IFNβ on ferroptosis in vivo. HT1080 cells, three-dimensional (3D) spheroids, and the xenograft mouse models were treated with IFNβ, RSL3, or IFNβ combination with RSL3 to analyze whether IFNβ enhances RSL3-induced ferroptosis. Results Here, we found that IFNβ could promote intracellular Fe2+ and lipid peroxidation levels, and decrease GSH levels in tumor cells. RNA sequencing data revealed that IFNβ induced a transcriptomic disturbance in ferroptosis-related genes. Knockdown of tripartite motif-containing 22 (TRIM22) suppressed the levels of intracellular Fe2+ and lipid ROS. It also reduced heme oxygenase (HMOX1) protein levels and increased ferroptosis suppressor protein 1 (FSP1) levels in HT1080 cells treated with IFNβ. Furthermore, our results illustrated that IFNβ enhanced the RAS-selective lethal 3 (RSL3)-induced ferroptosis and the inhibitory effect of RSL3 on GPX4. Meanwhile, compared to the groups treated with either IFNβ or RSL3 alone, the combination treatment of IFNβ and RSL3 significantly inhibited the growth of HT1080 three-dimensional (3D) spheroids and tumor in a mouse xenograft model. Conclusions Our work reveals a role for IFNβ in promoting ferroptosis and provides evidence that IFNβ could be used with RSL3 to increase cytotoxic effects in tumor cells.
Collapse
Affiliation(s)
- Huiyue Dong
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Ling Zhu
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jingjing Sun
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Clinical Laboratory, Wuhan Children’s Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyan Chen
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Pengyang Liu
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wei Zhang
- Fuzong Teaching Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Huajing Zeng
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Rong Lin
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Zongyang Yu
- Fuzong Teaching Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Pulmonary and Critical Care Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
- Fuzong Teaching Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Organ transplant institute, Dongfang Hospital, Xiamen University, Fuzhou, China
| |
Collapse
|
11
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2025; 480:759-784. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
12
|
Zhou Y, Wan Z, Xiong D, Gong Z, Liu F. Nur77 Promotes Inflammation in Cisplatin-Induced Acute Kidney Injury Through Transactivation of SERPINA3 Mediating Wnt/β-Catenin Pathway. Nephrology (Carlton) 2025; 30:e70006. [PMID: 39957271 DOI: 10.1111/nep.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/16/2025] [Accepted: 02/01/2025] [Indexed: 02/18/2025]
Abstract
AIM Acute kidney injury (AKI) is the most common complication in the treatment of cisplatin, which is a clinically effective and classical anticancer drug. Orphan Nuclear Receptor Nur77 has been found to promote renal ischaemia-reperfusion injury. In this study, we aim to explore the effects of Nur77 on cisplatin-induced AKI (CI-AKI) and its underlying mechanism. METHODS HK-2 cells treated with cisplatin were used to construct the CI-AKI model in vitro. Cell viability and cell proliferation were analysed using CCK-8 and EdU assays, respectively. Cell apoptosis was analysed by flow cytometry. The inflammation release level was detected using ELISA. Molecular abundance was evaluated using qPCR, Western blot and immunofluorescence. The interaction between Nur77 and SERPINA3 was clarified using ChIP and dual-luciferase reporter gene assays. RESULTS Our works demonstrated that Nur77 and SERPINA3 expression were considerably ascended in cisplatin-induced HK-2 cells. The silence of SERPINA3 alleviated cisplatin-stimulated HK-2 cell injury, which was characterised by increased cell viability and proliferation, and decreased apoptosis and inflammatory cytokine release. In addition, Nur77 promotes SERPINA3 transcription by binding to the SERPINA3 promoter region (-182 to -175), thereby upregulating SERPINA3 expression and activating the Wnt/β-catenin pathway. Moreover, HK-2 cell injury induced by cisplatin was notably inhibited by the knockdown of Nur77. Furthermore, the efficacy of Nur77 downregulation on the cell injury in cisplatin-stimulated HK-2 cells was antagonised by SERPINA3 overexpression. CONCLUSION Taken together, our findings revealed that Nur77 knockdown resisted cisplatin-induced HK-2 cells injury through lessening the expression of SERPINA3 mediated by transcriptional regulation and inactivating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Ying Zhou
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| | - Zhen Wan
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| | - Di Xiong
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| | - Zhijun Gong
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| | - Feiyan Liu
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| |
Collapse
|
13
|
Zheng F, Lei JZ, Wang JX, Xu XY, Zhou B, Ge R, Dai M, Dong HK, Wu N, Li YH, Zhu GQ, Zhou YB. Crucial roles of asprosin in cisplatin-induced ferroptosis and acute kidney injury. Free Radic Biol Med 2025; 227:296-311. [PMID: 39653130 DOI: 10.1016/j.freeradbiomed.2024.12.024] [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: 06/09/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Ferroptosis is a type of non-apoptotic regulated cell death characterized by iron accumulation and lipid peroxidation. Cisplatin is an effective chemotherapy drug with several serious side effects including acute kidney injury (AKI). Asprosin is a peptide contributing to metabolism regulation and metabolic disorders. This study aimed to determine the role and mechanism of asprosin in AKI. Cisplatin was used to induce cell damage in mouse renal tubular epithelial (TCMK-1) cells and AKI in C57BL/6 mice. Cisplatin caused asprosin upregulation in cisplatin-treated TCMK-1 cells and mice. In TCMK-1 cells, asprosin overexpression led to iron overload and lipid peroxidation, while asprosin knockdown attenuated cisplatin-induced iron overload, lipid peroxidation and ferroptosis. Exogenous asprosin promoted cell damage and ferroptosis, which were attenuated by ferroptosis inhibitors. Asprosin-induced iron overload, lipid peroxidation, cell damage and SMAD1/5/8 phosphorylation were prevented by bone morphogenetic protein (BMP) type I receptor inhibitor. Integrin antagonist prevented asprosin-induced SMAD1/5/8 phosphorylation, and asprosin can specifically bind to integrin β3. Inhibition of integrin β3 reduced the asprosin-induced increases in Fe2+ and MDA levels. Asprosin knockdown relieved cisplatin-induced hepcidin upregulation, while hepcidin knockdown attenuated asprosin-induced iron overload, lipid peroxidation and ferroptosis. In cisplatin-induced AKI mice, specific knockdown of asprosin in the kidney not only attenuated renal dysfunction and damage, but also alleviated iron overload, lipid peroxidation and ferroptosis. These results indicated that excessively increased asprosin promotes TCMK-1 cells ferroptosis and damage via integrin β3/BMP/hepcidin-mediated iron overload and lipid peroxidation. Silencing of asprosin attenuates renal injury and dysfunction in cisplatin-induced AKI by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Fen Zheng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jian-Zhen Lei
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jing-Xiao Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiao-Yu Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Bing Zhou
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, 241001, China
| | - Rui Ge
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Min Dai
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hong-Ke Dong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Nan Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Ye-Bo Zhou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
14
|
Zhang R, Wang J, Wu C, Wang L, Liu P, Li P. Lipidomics-based natural products for chronic kidney disease treatment. Heliyon 2025; 11:e41620. [PMID: 39866478 PMCID: PMC11758422 DOI: 10.1016/j.heliyon.2024.e41620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/17/2024] [Accepted: 12/31/2024] [Indexed: 01/28/2025] Open
Abstract
Chronic kidney disease (CKD) is by far the most prevalent disease in the world and is now a major global public health problem because of the increase in diabetes, hypertension and obesity. Traditional biomarkers of kidney function lack sensitivity and specificity for early detection and monitoring of CKD progression, necessitating more sensitive biomarkers for early diagnostic intervention. Dyslipidemia is a hallmark of CKD. Advancements in mass spectrometry (MS)-based lipidomics platforms have facilitated comprehensive analysis of lipids in biological samples and have revealed changes in the lipidome that are associated with metabolic disorders, which can be used as new biomarkers for kidney diseases. It is also critical for the discovery of new therapeutic targets and drugs. In this article, we focus on lipids in CKD, lipidomics methodologies and their applications in CKD. Additionally, we introduce novel biomarkers identified through lipidomics approaches and natural products derived from lipidomics for the treatment of CKD. We believe that our study makes a significant contribution to literature by demonstrating that natural products can improve CKD from a lipidomic perspective.
Collapse
Affiliation(s)
- Rui Zhang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Jingjing Wang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Chenguang Wu
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Lifan Wang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Peng Liu
- Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
15
|
Yang S, Chen L, Din S, Ye Z, Zhou X, Cheng F, Li W. The SIRT6/BAP1/xCT signaling axis mediates ferroptosis in cisplatin-induced AKI. Cell Signal 2025; 125:111479. [PMID: 39455033 DOI: 10.1016/j.cellsig.2024.111479] [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: 09/06/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Cisplatin is extensively utilized in clinical settings for treating solid tumors; However, its use is restricted because of the kidney damage caused by side effects. Moreover, currently, no effective medications have been approved to prevent or treat acute kidney injury induced by cisplatin. Our research indicates that sirtuin 6 (SIRT6) can inhibit ferroptosis induced by cisplatin, and the use of SIRT6 agonists can alleviate acute kidney injury caused by cisplatin. METHODS An animal model of cisplatin-induced acute kidney injury (AKI) was established, followed by RNA sequencing to identify potential differentially expressed genes (DEGs) and associated pathways. To explore the role of SIRT6 in this model, SIRT6 knockout mice were generated, and recombinant adeno-associated virus was employed to achieve SIRT6 overexpression in the mice. In vitro, cells were cultured in a cisplatin-containing medium to establish a cisplatin-induced cell model. The function of SIRT6 was further investigated by overexpressing or knocking down the gene using lentiviral plasmids. To elucidate the underlying molecular mechanisms, we employed RNA sequencing, performed bioinformatics analyses, and conducted chromatin immunoprecipitation assays. RESULTS RNA sequencing and Western blot analyses revealed a significant reduction in SIRT6 expression in mice with cisplatin-induced acute kidney injury (AKI). Enhancing SIRT6 expression improved renal function, reduced ferroptosis, and mitigated kidney damage, whereas SIRT6 knockout exacerbated kidney injury and heightened ferroptosis. Mechanistically, RNA sequencing, bioinformatics analysis, and chromatin immunoprecipitation assays demonstrated that SIRT6 inhibits ferroptosis by reducing the acetylation of histone H4K9ac at the BAP1 promoter. Furthermore, in vitro studies demonstrated that the SIRT6 agonist UBCS039 can alleviate cisplatin-induced acute kidney injury, highlighting its potential therapeutic role in mitigating cisplatin's damaging effects. However, further research is needed to fully elucidate the underlying mechanisms and to validate these findings in vivo. CONCLUSION Our findings underscore the critical role of the SIRT6/BAP1/xCT axis in regulating ferroptosis, particularly via the downregulation of SIRT6, in the context of cisplatin-induced acute kidney injury (AKI). This suggests that SIRT6 could be a promising therapeutic target for treating cisplatin-induced AKI. However, additional research is required to explore the specific mechanisms and fully assess the therapeutic potential of SIRT6 in this context.
Collapse
Affiliation(s)
- Songyuan Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lijia Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shikuan Din
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zehua Ye
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
16
|
Guo F, Wu Y, Wang G, Liu J. Role of PCBP2 in regulating nanovesicles loaded with curcumin to mitigate neuroferroptosis in neural damage caused by heat stroke. J Nanobiotechnology 2024; 22:800. [PMID: 39731111 DOI: 10.1186/s12951-024-02889-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/01/2024] [Indexed: 12/29/2024] Open
Abstract
OBJECTIVE This study aims to elucidate the mechanisms by which nanovesicles (NVs) transport curcumin(CUR) across the blood-brain barrier to treat hypothalamic neural damage induced by heat stroke by regulating the expression of poly(c)-binding protein 2 (PCBP2). METHODS Initially, NVs were prepared from macrophages using a continuous extrusion method. Subsequently, CUR was loaded into NVs using sonication, yielding engineered cell membrane Nanovesicles loaded with curcumin (NVs-CUR), which were characterized and subjected to in vitro and in vivo tracking analysis. Evaluations included assessing the toxicity of NVs-CUR using the MTT assay, evaluating neuroprotection of NVs-CUR against H2O2-induced oxidative stress damage in PC12 cells, examining effects on cell morphology and quantity, and detecting ferroptosis-related markers through Western blot and transmission electron microscopy (TEM). Proteomic analysis was conducted on PC12 cells treated with NVs (n = 3) and NVs-CUR (n = 3) to identify downstream key factors. Subsequently, the expression of key factors was modulated, and rescue experiments were performed to validate the impact of NVs-CUR through the regulation of key factor expression. Furthermore, a mouse model of hypothalamic neural damage induced by heat stroke was established, where CUR, NVs-CUR, and ferroptosis inducer Erastin were administered to observe mouse survival rates, conduct nerve function deficit scoring, perform histological staining, and measure levels of inflammation and oxidative stress factors in hypothalamic tissue. RESULTS NVs-CUR was successfully prepared with excellent stability, serving as an advantageous drug delivery system that effectively targets brain injury sites or neurons both in vitro and in vivo. Subsequent in vitro cell experiments demonstrated the biocompatibility of NVs-CUR, showing superior protective effects against H2O2-induced PC12 cell damage and reduced ferroptosis compared to CUR. Moreover, in the mouse model of hypothalamic neural damage induced by heat stroke, NVs-CUR exhibited enhanced therapeutic effects. Proteomic analysis revealed that NVs-CUR exerted its effects through the regulation of key protein PCBP2; silencing PCBP2 reversed the protective effect of NVs-CUR on neural damage and its inhibition of ferroptosis. Additionally, NVs-CUR regulated solute carrier family 7 member 11 (SLC7A11) expression by PCBP2; overexpression of SLC7A11 reversed the promotion of neural damage and ferroptosis by silencing PCBP2. Animal experiments indicated that ferroptosis inducers reversed the improved survival and nerve function observed with NVs-CUR, silencing PCBP2 reversed the ameliorative effects of NVs-CUR on hypothalamic neural injury induced by heat stroke, and overexpression of SLC7A11 further reversed the adverse effects of silencing PCBP2 on hypothalamic neural injury induced by heat stroke. This suggests that NVs-CUR alleviates hypothalamic neural damage induced by heat stroke by targeting the PCBP2/SLC7A11 axis to reduce neuronal ferroptosis. CONCLUSION This study successfully developed engineered cell membrane NVs-CUR with neuron-targeting properties. NVs-CUR increased the expression of PCBP2, maintained the stability of SLC7A11 mRNA, reduced ferroptosis, and ultimately alleviated hypothalamic neuroinflammation induced by heatstroke.
Collapse
Affiliation(s)
- Fei Guo
- Department of Emergency Trauma Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yizhan Wu
- Graduate School of Xinjiang Medical University, Urumqi, China
| | - Guangjun Wang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, No. 359, Youhao North Road, Urumqi, Xinjiang, China
| | - Jiangwei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, No. 359, Youhao North Road, Urumqi, Xinjiang, China.
| |
Collapse
|
17
|
Wang Y, Lv W, Ma X, Diao R, Luo X, Shen Q, Xu M, Yin M, Jin Y. NDUFS3 alleviates oxidative stress and ferroptosis in sepsis induced acute kidney injury through AMPK pathway. Int Immunopharmacol 2024; 143:113393. [PMID: 39426231 DOI: 10.1016/j.intimp.2024.113393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
In recent years, ferroptosis has been found to play an important role in various acute kidney injury (AKI). However, relatively little research has been conducted on sepsis-induced acute kidney injury (SI-AKI). As an important trigger of ferroptosis, how mitochondrial damage plays a regulatory role in SI-AKI is still unclear. To explore the potential relationship between mitochondria and ferroptosis, we established a SI-AKI rat model by intraperitoneal injection of lipopolysaccharide (LPS). Transcriptome sequencing was used to detect changes in gene transcription levels in the control group, LPS 3 h group, LPS 6 h group and LPS 12 h group. The severity of kidney injury was determined based on serum creatinine (CRE), blood urea nitrogen (BUN), tissue HE staining, TUNEL staining and inflammatory factor levels. Cytoscape software was utilized to screen several mitochondria-related HUB genes, and NADH dehydrogenase [ubiquinone] ferrithionein 3 (NDUFS3) was selected for subsequent validation due to its novelty and feasibility. qRT-PCR, Western blot was employed to evaluate the expression of NDUFS3 in kidney tissues. GO enrichment analysis revealed that up-regulated genes in the LPS 12 h group were enriched in several cell death terms while down-regulated genes were enriched in lipid metabolic process and oxidation-reduction progress terms. Furthermore, Western blot, IHC, MDA, GSH and iron content levels were used to assess ferroptosis in the kidney tissue of the SI-AKI rats, dihydroethidium (DHE) assay and ATP kit were used to assess mitochondrial ROS levels and mitochondrial function. To further validate the function of NDUFS3, we constructed overexpression rats using hydrodynamic method by tail vein injection of pc DNA3.1-NDUFS3 overexpression plasmid. we utilized LPS to stimulate HK-2 cells and establish an in vitro model. We then overexpressed NDUFS3 using pcDNA 3.1. The overexpression of NDUFS3 was found to inhibit LPS-induced ferroptosis and mitochondrial damage in HK-2 cells, as evidenced by Western blot, MDA, GSH, divalent iron, ROS levels, Mitosox red, ATP content and transmission electron microscopy. Finally, the use of Compound C to inhibit AMPK in HK-2 cells demonstrated that NDUFS3 plays a protective role through the AMPK pathway. Therefore, our study supports the emerging role of NDUFS3 in SI-AKI, providing new potential mitochondria-related targets for the treatment of SI-AKI.
Collapse
Affiliation(s)
- YuChen Wang
- Department of Laboratory Diagnosis, The First Afliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, People's Republic of China
| | - WuYang Lv
- Department of Laboratory Diagnosis, The First Afliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, People's Republic of China; Department of Laboratory Diagnosis, Shangluo Central Hospital, 148 Beixin Street, Shangluo 726099, Shaanxi, People's Republic of China
| | - XiaoTong Ma
- Department of Laboratory Diagnosis, The First Afliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, People's Republic of China
| | - RuXue Diao
- Department of Laboratory Diagnosis, The First Afliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, People's Republic of China
| | - XiaoXiao Luo
- Department of Laboratory Diagnosis, The First Afliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, People's Republic of China
| | - QiuLing Shen
- Department of Laboratory Diagnosis, The First Afliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, People's Republic of China
| | - MingYu Xu
- Department of Laboratory Diagnosis, The First Afliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, People's Republic of China
| | - MengJiao Yin
- Department of Laboratory Diagnosis, The First Afliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, People's Republic of China
| | - YingYu Jin
- Department of Laboratory Diagnosis, The First Afliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
18
|
Bingül İ, Kalayci R, Tekkeşin MS, Olgac V, Bekpinar S, Uysal M. Chenodeoxycholic acid alleviated the cyclosporine-induced nephrotoxicity by decreasing oxidative stress and suppressing renin-angiotensin system through AT2R and ACE2 mRNA upregulation in rats. J Mol Histol 2024; 56:23. [PMID: 39627449 DOI: 10.1007/s10735-024-10308-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: 06/25/2024] [Accepted: 10/04/2024] [Indexed: 02/07/2025]
Abstract
Oxidative stress, inflammation and renin-angiotensin system (RAS) activation play an important role in the nephrotoxicity which is caused by the long-term use of the immunosuppressive drug cyclosporine (CsA). This study investigates whether chenodeoxycholic acid (CDCA), an endogenous farnesoid X receptor (FXR) agonist with antioxidant and anti-inflammatory effects, modulates CsA nephrotoxicity. CsA (25 mg/kg/day; s.c.) was administered to rats for 12 days. CDCA (20 mg/kg/day; i.p.) injection was started 3 days before CsA and continued for 15 days. CDCA improved renal damage and function in CsA-administered rats. Renal function markers in serum, renal histology, oxidative stress, inflammation and RAS components were determined in kidney. CDCA reduced CsA-induced renal increases in NADPH oxidase 4 and NADPH oxidase 2 mRNA expressions, oxidative stress and inflammation. CDCA elevated renal FXR, small heterodimer partner-1, hypoxia-inducible factor and vascular endothelial growth factor and nuclear factor erythroid 2-related factor mRNA expressions in CsA rats. It prevents renin angiotensin system activation by reducing angiotensin II (Ang-II) levels in serum and upregulating renal mRNA expressions of Ang II type-II receptor (AT2R) and angiotensin converting enzyme 2 (ACE2), but not AT1R and ACE in CsA rats. Our results indicate that CDCA may be a protective agent against CsA-nephrotoxicity by decreasing inflammation, oxidative stress and RAS activation via AT2R and ACE2 upregulations.
Collapse
Affiliation(s)
- İlknur Bingül
- Department of Medical Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
| | - Rivaze Kalayci
- Department of Laboratory Animal Science Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Merva Soluk Tekkeşin
- Department of Oral Pathology, Faculty of Dentistry, Istanbul University, Istanbul, Turkey
| | - Vakur Olgac
- Department of Oral Pathology, Faculty of Dentistry, Istanbul University, Istanbul, Turkey
| | - Seldag Bekpinar
- Department of Medical Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mujdat Uysal
- Department of Medical Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
19
|
Zhang X, Wu W, Li Y, Peng Z. Exploring the role and therapeutic potential of lipid metabolism in acute kidney injury. Ren Fail 2024; 46:2403652. [PMID: 39319697 PMCID: PMC11425701 DOI: 10.1080/0886022x.2024.2403652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Acute kidney injury (AKI) is a prevalent condition, yet no specific treatment is available. Extensive research has revealed the pivotal role of lipid-related alterations in AKI. Lipid metabolism plays an essential role in the sustenance of the kidneys. In addition to their energy-supplying function, lipids contribute to the formation of renal biomembranes and the establishment of the renal microenvironment. Moreover, lipids or their metabolites actively participate in signal transduction, which governs various vital biological processes, such as proliferation, differentiation, apoptosis, autophagy, and epithelial-mesenchymal transition. While previous studies have focused predominantly on abnormalities in lipid metabolism in chronic kidney disease, this review focuses on lipid metabolism anomalies in AKI. We explore the significance of lipid metabolism products as potential biomarkers for the early diagnosis and classification of AKI. Additionally, this review assesses current preclinical investigations on the modulation of lipid metabolism in the progression of AKI. Finally, on the basis of existing research, this review proposes future directions, highlights challenges, and presents novel targets and innovative ideas for the treatment of and intervention in AKI.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Wen Wu
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
- Department of Critical Care Medicine, Center of Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Wu Q, Huang F. Targeting ferroptosis as a prospective therapeutic approach for diabetic nephropathy. Ann Med 2024; 56:2346543. [PMID: 38657163 PMCID: PMC11044758 DOI: 10.1080/07853890.2024.2346543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Diabetic nephropathy (DN) is a severe complication of diabetes mellitus, causing a substantive threat to the public, which receives global concern. However, there are limited drugs targeting the treatment of DN. Owing to this, it is highly crucial to investigate the pathogenesis and potential therapeutic targets of DN. The process of ferroptosis is a type of regulated cell death (RCD) involving the presence of iron, distinct from autophagy, apoptosis, and pyroptosis. A primary mechanism of ferroptosis is associated with iron metabolism, lipid metabolism, and the accumulation of ROS. Recently, many studies testified to the significance of ferroptosis in kidney tissue under diabetic conditions and explored the drugs targeting ferroptosis in DN therapy. Our review summarized the most current studies between ferroptosis and DN, along with investigating the significant processes of ferroptosis in different kidney cells, providing a novel target treatment option for DN.
Collapse
Affiliation(s)
- Qinrui Wu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fengjuan Huang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
21
|
Famurewa AC, George MY, Ukwubile CA, Kumar S, Kamal MV, Belle VS, Othman EM, Pai SRK. Trace elements and metal nanoparticles: mechanistic approaches to mitigating chemotherapy-induced toxicity-a review of literature evidence. Biometals 2024; 37:1325-1378. [PMID: 39347848 DOI: 10.1007/s10534-024-00637-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
Anticancer chemotherapy (ACT) remains a cornerstone in cancer treatment, despite significant advances in pharmacology over recent decades. However, its associated side effect toxicity continues to pose a major concern for both oncology clinicians and patients, significantly impacting treatment protocols and patient quality of life. Current clinical strategies to mitigate ACT-induced toxicity have proven largely unsatisfactory, leaving a critical unmet need to block toxicity mechanisms without diminishing ACT's therapeutic efficacy. This review aims to document the molecular mechanisms underlying ACT toxicity and highlight research efforts exploring the protective effects of trace elements (TEs) and their nanoparticles (NPs) against these mechanisms. Our literature review reveals that the primary driver of ACT toxicity is redox imbalance, which triggers oxidative inflammation, apoptosis, endoplasmic reticulum stress, mitochondrial dysfunction, autophagy, and dysregulation of signaling pathways such as PI3K/mTOR/Akt. Studies suggest that TEs, including zinc, selenium, boron, manganese, and molybdenum, and their NPs, can potentially counteract ACT-induced toxicity by inhibiting oxidative stress-mediated pathways, including NF-κB/TLR4/MAPK/NLRP3, STAT-3/NLRP3, Bcl-2/Bid/p53/caspases, and LC3/Beclin-1/CHOP/ATG6, while also upregulating protective signaling pathways like Sirt1/PPAR-γ/PGC-1α/FOXO-3 and Nrf2/HO-1/ARE. However, evidence regarding the roles of lncRNA and the Wnt/β-catenin pathway in ACT toxicity remains inconsistent, and the impact of TEs and NPs on ACT efficacy is not fully understood. Further research is needed to confirm the protective effects of TEs and their NPs against ACT toxicity in cancer patients. In summary, TEs and their NPs present a promising avenue as adjuvant agents for preventing non-target organ toxicity induced by ACT.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike Ikwo, Abakaliki, Ebonyi, Nigeria.
- Centre for Natural Products Discovery, School of P harmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Cletus A Ukwubile
- Department of Pharmacognosy, Faculty of Pharmacy, University of Maiduguri, Bama Road, Maiduguri, Borno, Nigeria
| | - Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mehta V Kamal
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vijetha S Belle
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Eman M Othman
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
- Cancer Therapy Research Center, Department of Biochemistry-I, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
- Department of Bioinformatics, University of Würzburg, Am Hubland, 97074, BiocenterWürzburg, Germany
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
22
|
Deng M, Tan X, Peng X, Zheng W, Fu R, Tao S. HDAC6 promotes inflammation in lupus nephritis mice by regulating transcription factors MAFF and KLF5 in renal fibrosis. Ren Fail 2024; 46:2415517. [PMID: 39412062 PMCID: PMC11485742 DOI: 10.1080/0886022x.2024.2415517] [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: 11/28/2023] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
AIM This study explored the effect and mechanism of MAFF and HDAC6 on renal fibrosis and inflammation in lupus nephritis (LN). METHODS IL-33 treated renal epithelial cells and MRL/lpr mice were respectively used for in vitro and in vivo experiments. The expressions of HDAC6, MAFF, and KLF5 were measured in cells and renal tissues. Before and after cell transfection, the morphological changes in renal tissues were observed using Hematoxylin and eosin (H&E) and Masson staining. The proteinuria, serum creatinine (SCr), blood urea nitrogen (BUN), and double-stranded DNA (dsDNA) levels were detected by biochemical analysis. The expressions of fibrosis and inflammation related proteins (including α-SMA, Vimentin, IL-1β, IL-6, and TNF-α), p65, and iNOS were also detected. The relationship among MAFF, HDAC6, and KLF5 was determined by chromatin immunoprecipitation and dual luciferase reporter gene assay. RESULTS Renal tissues and cell models had elevated expressions of HDAC6 and KLF5, and decreased MAFF expression. HDAC6 suppression or MAFF overexpression led to suppression of proteinuria, SCr, BUN, and dsDNA levels, as well as attenuation of inflammatory infiltration and collagen deposition. HDAC6 can suppress MAFF expression via deacetylation to abolish its suppression of KLF5 expression, thus increasing KLF5 expression. In vivo and in vitro experiments showed the suppressive effect of HDAC6 suppression on renal fibrosis and inflammation can be abolished by KLF5 overexpression. CONCLUSION HDAC6 suppresses MAFF expression via deacetylation to elevate KLF5 expression, which consequently enhances fibrosis and inflammatory response in LN.
Collapse
Affiliation(s)
- Meihui Deng
- Department of Nephrology, Jiangxi Provincial Children’s Hospital, Nanchang, Jiangxi, P.R. China
| | - Xiao Tan
- Department of Hematology, Jiangxi Provincial Children’s Hospital, Nanchang, Jiangxi, P.R. China
| | - Xiaojie Peng
- Department of Nephrology, Jiangxi Provincial Children’s Hospital, Nanchang, Jiangxi, P.R. China
| | - Weimin Zheng
- Department of Nephrology, Jiangxi Provincial Children’s Hospital, Nanchang, Jiangxi, P.R. China
| | - Rui Fu
- Department of Nephrology, Jiangxi Provincial Children’s Hospital, Nanchang, Jiangxi, P.R. China
| | - Shanshan Tao
- Department of Nephrology, Jiangxi Provincial Children’s Hospital, Nanchang, Jiangxi, P.R. China
| |
Collapse
|
23
|
Zhu J, Yuan A, Le Y, Chen X, Guo J, Liu J, Chen H, Wang CY, Lu D, Lu K. Yi-Qi-Jian-Pi-Xiao-Yu formula inhibits cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated ferritinophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156189. [PMID: 39515100 DOI: 10.1016/j.phymed.2024.156189] [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: 06/04/2024] [Revised: 09/28/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The kidneys are the primary excretory organs for platinum drugs, making them susceptible to damage from these drugs. Cisplatin-induced acute kidney injury (CIAKI) is the most common side effect observed in patients undergoing clinical cisplatin treatment. A traditional Chinese medicinal preparation, the Yi-Qi-Jian-Pi-Xiao-Yu formula (YQJPXY), which is a modified formulation of the classical Chinese medicine formula Buyang Huanwu Decoction, has long been used in the treatment of clinical kidney diseases. It is expected to be used to ameliorate cisplatin-induced acute kidney injury. However, the mechanism of this YQJPXY for the treatment of cisplatin-induced acute kidney injury remains unclear. PURPOSE The objective of this study is to examine the impact of the YQJPXY on the inhibition of ferroptosis in cisplatin-induced acute kidney injury and to elucidate the underlying mechanisms. METHODS The active components of YQJPXY were analysed using UPLC-MS/MS. A comprehensive investigation was conducted to elucidate the effects and regulatory mechanisms of YQJPXY on CIAKI and ferroptosis in mice subjected to acute cisplatin treatment and in mice receiving cisplatin treatment after STING expression was inhibited using the STING inhibitor C176. The renoprotective effect of YQJPXY on cisplatin-treated mice was evaluated by measuring tissue damage, inflammation and pro-fibrosis. In addition, we employed network pharmacology and molecular docking methodologies to analyse the principal regulatory targets of YQJPXY. Furthermore, the expression of key proteins and markers of ferroptosis and iron metabolism, as well as the levels of key indicators related to STING-associated ferritinophagy, were examined by immunoblotting, immunohistochemistry, immunoprecipitation, quantitative real-time PCR (qPCR) and specific probes. RESULTS The results demonstrated that YQJPXY reduced the levels of indicators of injury, inflammation and pro-fibrosis in CIAKI mice, with renoprotective effects. Network pharmacological analyses revealed that ferroptosis might be the main biological process regulated by YQJPXY. Furthermore, molecular docking results indicated that STING might be a potential regulatory target of YQJPXY. Furthermore, YQJPXY treatment resulted in a significant reduction in MDA and 4-HNE levels, as well as the inhibition of ferroptosis and improvement in iron metabolic processes. Concomitantly, YQJPXY exhibited a robust protective effect on ferroptosis and iron metabolism homeostasis, as evidenced by its inhibitory action on ferritinophagy. Validation experiments utilising the cisplatin inhibitor C176 demonstrated that YQJPXY inhibits cisplatin-induced ferroptosis in kidney via STING-mediated ferritinophagy. CONCLUSION These suggest that YQJPXY alleviates cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated Ferritinophagy.
Collapse
Affiliation(s)
- Ji Zhu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou 330061, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Aini Yuan
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yifei Le
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaohui Chen
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Jianan Guo
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jing Liu
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hang Chen
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Cai-Yi Wang
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Dezhao Lu
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Keda Lu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou 330061, China.
| |
Collapse
|
24
|
Zheng ZL, Ma JW, Luo Y, Liang GJ, Lei SJ, Yan KJ, Meng HB, Liu XJ. Mechanism of dexmedetomidine protection against cisplatin induced acute kidney injury in rats. Ren Fail 2024; 46:2337287. [PMID: 38627212 PMCID: PMC11022910 DOI: 10.1080/0886022x.2024.2337287] [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: 09/26/2023] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE This study explored the molecular mechanisms by which dexmedetomidine (Dex) alleviates cisplatin (CP)-induced acute kidney injury (AKI) in rats. METHODS CP-induced AKI models were established, and Dex was intraperitoneally injected at different concentrations into rats in the model groups. Subsequently, rats were assigned to the control, CP, CP + Dex 10 μg/kg, and CP + Dex 25 μg/kg groups. After weighing the kidneys of the rats, the kidney arterial resistive index was calculated, and CP-induced AKI was evaluated. In addition, four serum biochemical indices were measured: histopathological damage in rat kidneys was detected; levels of inflammatory factors, interleukin (IL)-1β, IL-18, IL-6, and tumor necrosis factor alpha, in kidney tissue homogenate of rats were assessed through enzyme-linked immunosorbent assay (ELISA); and levels of NLRP-3, caspase-1, cleaved caspase-1, gasdermin D (GSDMD), and GSDMD-N in kidney tissues of rats were determined via western blotting. RESULTS Dex treatment reduced nephromegaly and serum clinical marker upregulation caused by CP-induced AKI. In addition, hematoxylin and eosin staining revealed that Dex treatment relieved CP-induced kidney tissue injury in AKI rats. ELISA analyses demonstrated that Dex treatment reduced the upregulated levels of proinflammatory cytokines in the kidney tissue of AKI rats induced by CP, thereby alleviating kidney tissue injury. Western blotting indicated that Dex alleviated CP-induced AKI by inhibiting pyroptosis mediated by NLRP-3 and caspase-1. CONCLUSION Dex protected rats from CP-induced AKI, and the mechanism may be related to NLRP-3/Caspase-1-mediated pyroptosis.
Collapse
Affiliation(s)
- Zeng-lu Zheng
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Jun-wei Ma
- Department of Nephrology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Yi Luo
- Department of Respiratory, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Gui-jin Liang
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Shi-jie Lei
- Department of Proctology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Ke-jin Yan
- Department of Proctology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Hai-bing Meng
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Xiu-juan Liu
- Department of Nephrology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| |
Collapse
|
25
|
Chen L, Wang Q, Li T, Li L, Wang C, Xu B, Gong X. Exploring therapeutic mechanisms of Chuan Huang Fang-II in the treatment of acute kidney injury on chronic kidney disease patients from the perspective of lipidomics. Ren Fail 2024; 46:2356021. [PMID: 38785301 PMCID: PMC11132756 DOI: 10.1080/0886022x.2024.2356021] [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: 09/19/2023] [Accepted: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
OBJECTIVE This study aims to assess the clinical efficacy and safety of CHF-II in combination with RG for treating AKI on CKD (A on C), and to explore potential therapeutic mechanisms through lipidomics analysis. METHODS 98 patients were enrolled and randomly assigned to the RG or RG + CHF groups. Both groups received RG therapy, with RG + CHF group additionally receiving CHF-II treatment over a duration of two weeks. Evaluation endpoints included changes in renal function, blood lipid profiles, urinary AKI biomarkers, and TCM symptoms before and after treatment. Serum samples were collected for lipid metabolite analysis. RESULTS The total clinical effective rate in RG + CHF group was 73.5%, and that of RG group was 40.8%. TCM syndrome scores in RG + CHF group showed a more pronounced decrease (p < 0.05). Scr, BUN, and UA levels decreased while eGFR levels increased in both groups (p < 0.05), with a greater magnitude of change observed in the RG + CHF group. Urinary AKI biomarkers decreased more in RG + CHF group (p < 0.05). No serious adverse events occurred during the trial. 58 different lipid metabolites and 48 lipid biomarkers were identified. According to the KEGG database, the possible metabolic pathways involved triglyceride metabolic pathway and fat digestion and absorption metabolic pathways. CONCLUSION CHF-II effectively alleviated kidney injury and improved TCM syndrome scores in patients with A on C. Lipid differential metabolites could serve as diagnostic indicators for AKI in patients with CKD. The possible metabolic pathways might be implicated in therapeutic action of CHF-II in the prevention and treatment of patients with A on C.
Collapse
Affiliation(s)
- Ling Chen
- Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Wang
- Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tonglu Li
- Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lejia Li
- Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bing Xu
- Department of Nephrology, Minhang Branch of Yueyang Hospital of Integrative Chinese & Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuezhong Gong
- Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Zhou Q, Dian Y, He Y, Yao L, Su H, Meng Y, Sun Y, Li D, Xiong Y, Zeng F, Liang X, Liu H, Chen X, Deng G. Propafenone facilitates mitochondrial-associated ferroptosis and synergizes with immunotherapy in melanoma. J Immunother Cancer 2024; 12:e009805. [PMID: 39581704 PMCID: PMC11590812 DOI: 10.1136/jitc-2024-009805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Despite the successful application of immunotherapy, both innate and acquired resistance are typical in melanoma. Ferroptosis induction appears to be a potential strategy to enhance the effectiveness of immunotherapy. However, the relationship between the status of ferroptosis and the effectiveness of immunotherapy, as well as viable strategies to augment ferroptosis, remains unclear. METHODS A screening of 200 cardiovascular drugs obtained from the Food and Drug Administration-approved drug library was conducted to identify the potential ferroptosis sensitizer. In vitro and in vivo experiments explored the effects of propafenone on ferroptosis in melanoma. Animal models and transcriptomic analyses evaluated the therapeutic effects and survival benefits of propafenone combined with immune checkpoint blockades (ICBs). The relationship between propafenone targets and the efficacy of ICBs was validated using the Xiangya melanoma data set and publicly available clinical data sets. RESULTS Through large-scale drug screening of cardiovascular drugs, we identified propafenone, an anti-arrhythmia medication, as capable of synergizing with ferroptosis inducers in melanoma. Furthermore, we observed that propafenone, in combination with glutathione peroxidase 4 inhibitor RSL3, collaboratively induces mitochondrial-associated ferroptosis. Mechanistically, propafenone transcriptionally upregulates mitochondrial heme oxygenase 1 through the activation of the Jun N-terminal kinase (JNK)/JUN signaling pathway under RSL3 treatment, leading to overloaded ferrous iron and reactive oxygen species within the mitochondria. In xenograft models, the combination of propafenone and ferroptosis induction led to nearly complete tumor regression and prolonged survival. Consistently, propafenone enhances immunotherapy-induced tumorous ferroptosis and antitumor immunity in tumor-bearing mice. Significantly, patients exhibiting high levels of ferroptosis/JUN/HMOX1 exhibited improved efficacy of immunotherapy and prolonged progression-free survival. CONCLUSIONS Taken together, our findings suggest that propafenone holds promise as a candidate drug for enhancing the efficacy of immunotherapy and other ferroptosis-targeted therapies in the treatment of melanoma.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Yi He
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Lei Yao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hui Su
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Yixiao Xiong
- Department of Dermatology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiaowei Liang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan Province, China
- Furong Laboratory, Changsha, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| |
Collapse
|
27
|
Zhang S, Zhang D, Xu K, Huang X, Chen Q, Chen M. The role of the farnesoid X receptor in diabetes and its complications. Mol Cell Biochem 2024. [DOI: 10.1007/s11010-024-05162-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/10/2024] [Indexed: 01/06/2025]
|
28
|
Ye Y, Liang J, Xu C, Liu Y, Chen J, Zhu Y. Inhibition of HMOX1 by MAFG potentiates the development of depression‑like behavior in mice associated with astrocyte-mediated neuroinflammation. Brain Res 2024; 1843:149115. [PMID: 38977234 DOI: 10.1016/j.brainres.2024.149115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/23/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
MAF bZIP transcription factor G (MAFG)-driven astrocytes have been reported to promote inflammation in the CNS. However, its function in depression, the primary cause of disability worldwide, has not been well characterized. This study investigated the possible perturbation of heme oxygenase 1 (HMOX1, also known as HO1) by the transcription factor MAFG as an underlying mechanism of the development of depression. The GSE98793 dataset was included for gene expression analysis of whole blood from donors with major depressive disorder and controls, and the target of MAFG was predicted by multiple database mining. Mouse and cellular models of depression were established by chronic unpredictable mild stress (CUMS) and lipopolysaccharide (LPS) treatment of astrocytes, which were treated with MAFG and HMOX1 knockdown plasmids. MAFG was highly expressed in the hippocampal tissues of CUMS-challenged mice and LPS-induced astrocytes. MAFG knockdown alleviated depression-like behaviors in mice. MAFG bound to the HMOX1 promoter and repressed its transcription. Knockdown of HMOX1 exacerbated neuroinflammation in astrocytes and accelerated depression-like behavior in mice. In conclusion, MAFG knockdown attenuated CUMS-stimulated depression-like behaviors in mice by astrocyte-mediated neuroinflammation via restoration of HMOX1.
Collapse
Affiliation(s)
- Ying Ye
- Department of Psychiatry, The Seventh People's Hospital of Wenzhou, Wenzhou 325006, Zhejiang, PR China
| | - Jiawei Liang
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 318150, Zhejiang, PR China
| | - Cheng Xu
- Department of Pathology, Enze Hospital, Taizhou Enze Medical Center, Taizhou 318050, Zhejiang, PR China
| | - Yang Liu
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu 610000, Sichuan, PR China
| | - Jia Chen
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu 610000, Sichuan, PR China; The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Yanhui Zhu
- Department of Psychiatry, The Seventh People's Hospital of Wenzhou, Wenzhou 325006, Zhejiang, PR China.
| |
Collapse
|
29
|
Huang P, Zhao H, Dai H, Li J, Pan X, Pan W, Xia C, Liu F. FXR deficiency induced ferroptosis via modulation of the CBP-dependent p53 acetylation to suppress breast cancer growth and metastasis. Cell Death Dis 2024; 15:826. [PMID: 39543094 PMCID: PMC11564727 DOI: 10.1038/s41419-024-07222-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Farnesoid X receptor (NR1H4/FXR) functions as a scavenger of lipid peroxide products and drives the proliferation and metastasis of various cancers. However, the underlying molecular mechanisms remain poorly understood. In our study, we found that the expression levels of FXR, vimentin and SLC7A11 were significantly higher in breast cancer tissues, particularly in metastatic cancer tissues compared to non-metastatic ones. Furthermore, the increased FXR expression was positively correlated with vimentin and SLC7A11 in clinical tumor specimens. In addition, a high level of FXR correlated with poor prognosis in patients with breast cancer. Both Z-Guggulsterone (Z-GS), as a pharmacological inhibitor of FXR, and silencing FXR curbed proliferation and migration of breast cancer cells by promoting ferroptosis. Notably, our results showed that FXR competitively bound to CREB-binding protein (CBP) to suppress the interaction between p53 and CBP in the nucleus, and thus prevented p53 acetylation at lys382, which was essential for upregulating the expression of SLC7A11. Conversely, FXR knockdown increased the interaction between p53 and CBP and promoted p53 acetylation, which ultimately led to facilitating ferroptosis in breast cancer cells. More importantly, we also found that Z-GS inhibited TGF-β1-induced tumor growth and metastasis of breast cancer primarily through ferroptosis via regulating CBP-dependent p53 acetylation in nude mice. In conclusion, the FXR was first reported as a tumor promoter that enhanced the proliferation and metastasis of breast cancer cells through regulating CBP-dependent p53 K382 acetylation. It proposes that FXR may serve as a potential therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Ping Huang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Han Zhao
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Hua Dai
- Department of Pathology, the First Affiliated Hospital of Nanchang University, Nanchang, 330038, P. R. China
| | - Jinying Li
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Xiafang Pan
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Wentian Pan
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Chunhua Xia
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
- Jiangxi Province Key Laboratory of New Drug Evaluation and Transformation, Nanchang, 330031, P. R. China.
| | - Fanglan Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
- Jiangxi Province Key Laboratory of New Drug Evaluation and Transformation, Nanchang, 330031, P. R. China.
| |
Collapse
|
30
|
Zeng L, Huang J, Wang Y, Hu Y, Zhou S, Lu Y. Oleanolic acid induces hepatic injury by disrupting hepatocyte tight junction and dysregulation of farnesoid X receptor-mediated bile acid efflux transporters. J Appl Toxicol 2024; 44:1725-1741. [PMID: 39030772 DOI: 10.1002/jat.4667] [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: 12/06/2023] [Revised: 06/09/2024] [Accepted: 06/20/2024] [Indexed: 07/22/2024]
Abstract
Oleanolic acid (OA) is a naturally occurring pentacyclic triterpene compound that has been reported to cause cholestatic liver injury. However, the regulation and pathogenic role of bile acids in OA-induced development of cholestatic liver injury remains largely unclear. Farnesoid X receptor (FXR) is a metabolic nuclear receptor that plays an important role in bile acid homeostasis in the liver by regulating efflux transporters bile salt export pump (BSEP) and multidrug resistance-associated protein 2 (MRP2). The aim of this study was to investigate the effect of OA on hepatocyte tight junction function and determine the role of FXR, BSEP, and MRP2 in the mechanism of impairment of transport of bile acids induced by OA. Both in vivo and in vitro models were used to characterize the OA-induced liver injury. The liquid chromatography-tandem mass spectrometry (LC-MS) was employed to characterize the efflux function of the transporters, and the results showed that OA caused a blockage of bile acids efflux. OA treatment resulted in decreased expression levels of the tight junction proteins zonula occludens-1 and occludin. Immunofluorescence results showed that OA treatment significantly reduced the number of bile ducts and the immunofluorescence intensity. Pretreatment with agonists of FXR and MRP2, respectively, in animal experiments attenuated OA-induced liver injury, while pretreatment with inhibitors of BSEP and MRP2 further aggravated OA-induced liver injury. These results suggest that OA inhibits FXR-mediated BSEP and MRP2, leading to impaired bile acid efflux and disruption of tight junctions between liver cells, resulting in liver damage.
Collapse
Affiliation(s)
- Li Zeng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Jianxiang Huang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- School of Pharmacy, Zunyi Medical University, Zunyi, China
- Department of Pharmacy, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Yi Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yan Hu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Shaoyu Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yuanfu Lu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
31
|
Liang NN, Guo YY, Zhang XY, Ren YH, He YZ, Liu ZB, Xu DX, Xu S. Mitochondrial Dysfunction-Evoked DHODH Acetylation is Involved in Renal Cell Ferroptosis during Cisplatin-Induced Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404753. [PMID: 39303219 DOI: 10.1002/advs.202404753] [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: 05/02/2024] [Revised: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Several studies have observed renal cell ferroptosis during cisplatin-induced acute kidney injury (AKI). However, the mechanism is not completely clear. In this study, oxidized arachidonic acid (AA) metabolites are increased in cisplatin-treated HK-2 cells. Targeted metabolomics showed that the end product of pyrimidine biosynthesis is decreased and the initiating substrate of pyrimidine biosynthesis is increased in cisplatin-treated mouse kidneys. Mitochondrial DHODH, a key enzyme for pyrimidine synthesis, and its downstream product CoQH2, are downregulated. DHODH overexpression attenuated but DHODH silence exacerbated cisplatin-induced CoQH2 depletion and lipid peroxidation. Mechanistically, renal DHODH acetylation is elevated in cisplatin-exposed mice. Mitochondrial SIRT3 is reduced in cisplatin-treated mouse kidneys and HK-2 cells. Both in vitro SIRT3 overexpression and in vivo NMN supplementation attenuated cisplatin-induced mitochondrial DHODH acetylation and renal cell ferroptosis. By contrast, Sirt3 knockout aggravated cisplatin-induced mitochondrial DHODH acetylation and renal cell ferroptosis, which can not be attenuated by NMN. Additional experiments showed that cisplatin caused mitochondrial dysfunction and SIRT3 SUMOylation. Pretreatment with mitochondria-target antioxidant MitoQ alleviated cisplatin-caused mitochondrial dysfunction, SIRT3 SUMOylation, and DHODH acetylation. MitoQ pretreatment protected against cisplatin-caused AKI and renal cell ferroptosis. Taken together, these results suggest that mitochondrial dysfunction-evoked DHODH acetylation partially contributes to renal cell ferroptosis during cisplatin-induced AKI.
Collapse
Affiliation(s)
- Nan-Nan Liang
- Department of Toxicology, Anhui Medical University, Hefei, China, 230032
| | - Yue-Yue Guo
- Department of Toxicology, Anhui Medical University, Hefei, China, 230032
| | - Xiao-Yi Zhang
- Department of Toxicology, Anhui Medical University, Hefei, China, 230032
| | - Ya-Hui Ren
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China, 230601
| | - Yi-Zhang He
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China, 230601
| | - Zhi-Bing Liu
- Department of Blood Transfusion, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, China, 230032
| | - Shen Xu
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China, 230601
| |
Collapse
|
32
|
Gao Q, Chen JM, Li CSZ, Zhan JY, Yin XD, Li BS, Dong HL, Luo LX, Li ZL. CDKN1A promotes Cis-induced AKI by inducing cytoplasmic ROS production and ferroptosis. Food Chem Toxicol 2024; 193:115003. [PMID: 39353481 DOI: 10.1016/j.fct.2024.115003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND AND OBJECTIVE This study focuses on investigating the role of CDKN1A in cisplatin-induced AKI (acute kidney injury, AKI) and its potential as a biomarker for early diagnosis and therapeutic intervention by integrating bioinformatics analysis, machine learning, and experimental validation. METHODS We analyzed the GSE85957 dataset to find genes that changed between control and cisplatin-treated rats. Using bioinformatics and machine learning, we found 13 important genes related to ferroptosis and the P53 pathway. The key gene, CDKN1A, was identified using various algorithms. We then tested how reducing CDKN1A in human kidney cells affected cell health, ROS, and iron levels. We also checked how CDKN1A changes the levels of proteins linked to ferroptosis using Q-PCR and Western Blot. RESULTS CDKN1A was found to negatively regulate the G1/S phase transition and was associated with ferroptosis in p53 signaling. Experiments in human renal tubular epithelial cells (HK-2) and rat NRK-52E cells showed that CDKN1A knockdown mitigated cisplatin-induced cell injury by reducing oxidative stress and ferroptosis. CONCLUSION Our integrated approach identified CDKN1A as a biomarker for cisplatin-induced AKI. Its regulation could be key in AKI pathogenesis, offering new therapeutic insights and aiding in early diagnosis and intervention.
Collapse
Affiliation(s)
- Qian Gao
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Medicine, Shanghai Jiao Tong University, Shanghai, 200125, China
| | - Jun-Ming Chen
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Chen-Sui-Zi Li
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Medicine, Shanghai Jiao Tong University, Shanghai, 200125, China
| | - Jia-Yi Zhan
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Xue-Dong Yin
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Ben-Shang Li
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, China
| | - Hong-Liang Dong
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200120, China.
| | - Lian-Xiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| | - Zhi-Ling Li
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
33
|
Liu W, Xiong Z, Fu T, Yang J, Zou J, Wu Y, Kuang L, Wang Q, Li S, Le A. Regulation of renal ischemia-reperfusion injury and tubular epithelial cell ferroptosis by pparγ m6a methylation: mechanisms and therapeutic implications. Biol Direct 2024; 19:99. [PMID: 39444036 PMCID: PMC11515743 DOI: 10.1186/s13062-024-00515-9] [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: 03/18/2024] [Accepted: 08/08/2024] [Indexed: 10/25/2024] Open
Abstract
This study aimed to elucidate the role and underlying mechanisms of Peroxisome proliferator-activated receptor gamma (PPARγ) and its m6A methylation in renal ischemia-reperfusion (I/R) injury and ferroptosis of tubular epithelial cells (TECs). High-throughput transcriptome sequencing was performed on renal tissue samples from I/R injury models and sham-operated mice, complemented by in vivo and in vitro experiments focusing on the PPARγ activator Rosiglitazone and the manipulation of METTL14 and IGF2BP2 expression. Key evaluations included renal injury assessment, ferroptosis indicator measurement, and m6A methylation analysis of PPARγ. Our findings highlight the critical role of the PPARγ pathway and ferroptosis in renal I/R injury, with Rosiglitazone ameliorating renal damage and TEC ferroptosis. METTL14-mediated m6A methylation of PPARγ, dependent on IGF2BP2, emerged as a pivotal regulator of PPARγ expression, renal injury, and ferroptosis. This study reveals that PPARγ m6A methylation, orchestrated by METTL14 through an IGF2BP2-dependent mechanism, plays a crucial role in mitigating renal I/R injury and TEC ferroptosis. These insights offer promising avenues for therapeutic strategies targeting acute kidney injury.
Collapse
Affiliation(s)
- Wei Liu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Ziqing Xiong
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Tianmei Fu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Juan Yang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Juan Zou
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Yize Wu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Linju Kuang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Qian Wang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Song Li
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Aiping Le
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
34
|
Qin W, Huang J, Zhang M, Xu M, He J, Liu Q. Nanotechnology-Based Drug Delivery Systems for Treating Acute Kidney Injury. ACS Biomater Sci Eng 2024; 10:6078-6096. [PMID: 39226188 PMCID: PMC11480945 DOI: 10.1021/acsbiomaterials.4c01385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Acute kidney injury (AKI) is a disease that is characterized by a rapid decline in renal function and has a relatively high incidence in hospitalized patients. Sepsis, renal hypoperfusion, and nephrotoxic drug exposure are the main causes of AKI. The major therapy measures currently include supportive treatment, symptomatic treatment, and kidney transplantation. These methods are supportive treatments, and their results are not satisfactory. Fortunately, many new treatments that markedly improve the AKI therapy efficiency are emerging. These include antioxidant therapy, ferroptosis therapy, anti-inflammatory therapy, autophagy therapy, and antiapoptotic therapy. In addition, the development of nanotechnology has further promoted therapeutic effects on AKI. In this review, we highlight recent advances in the development of nanocarriers for AKI drug delivery. Emphasis has been placed on the latest developments in nanocarrier modification and design. We also summarize the applications of different nanocarriers in AKI treatment. Finally, the advantages and challenges of nanocarrier applications in AKI are summarized, and several nanomedicines that have been approved for clinical trials to treat diverse kidney diseases are listed.
Collapse
Affiliation(s)
- Wanbing Qin
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Jiaqi Huang
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Manting Zhang
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Mingwei Xu
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Junbing He
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Qinghua Liu
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
- Department
of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 Guangdong, China
- NHC Key
Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong
Provincial Key Laboratory of Nephrology, Guangzhou, 510080 Guangdong, China
| |
Collapse
|
35
|
Sharawy N, Aboulhoda B, Khalifa M, Morcos G, Morsy S, Alghamdi M, Khalifa I, Abd Algaleel W. Amelioration of nephrotoxicity by targeting ferroptosis: role of NCOA4, IREB2, and SLC7a11 signaling. Braz J Med Biol Res 2024; 57:e13116. [PMID: 39383377 PMCID: PMC11463912 DOI: 10.1590/1414-431x2024e13116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/29/2024] [Indexed: 10/11/2024] Open
Abstract
Nephrotoxicity is a common complication that limits the clinical utility of cisplatin. Ferroptosis is an iron-dependent necrotic cell death program that is mediated by phospholipid peroxidation. The molecular mechanisms that disrupt iron homeostasis and lead to ferroptosis are yet to be elucidated. In this study, we aimed to investigate the involvement of nuclear receptor coactivator 4 (NCOA4), a selective cargo receptor that mediates ferroptosis and autophagic degradation of ferritin in nephrotoxicity. Adult male Sprague-Dawley rats were randomly-assigned to four groups: control group, cisplatin (Cis)-treated group, deferiprone (DEF)-treated group, and Cis+DEF co-treated group. Serum, urine, and kidneys were isolated to perform biochemical, morphometric, and immunohistochemical analysis. Iron accumulation was found to predispose to ferroptotic damage of the renal tubular cells. Treatment with deferiprone highlights the role of ferroptosis in nephrotoxicity. Upregulation of NCOA4 in parallel with low ferritin level in renal tissue seems to participate in iron-induced ferroptosis. This study indicated that ferroptosis may participate in cisplatin-induced tubular cell death and nephrotoxicity through iron-mediated lipid peroxidation. Iron dyshomeostasis could be attributed to NCOA4-mediated ferritin degradation.
Collapse
Affiliation(s)
- N. Sharawy
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - B.E. Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - M.M. Khalifa
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Human Physiology, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - G.N. Morcos
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Basic Medical Sciences, Faculty of Medicine, King Salman International University, South Sinai, Sinai, Egypt
| | - S.A.A.G. Morsy
- Pathological Sciences Department, MBBS Program, Fakeeh College for Medical Sciences, Jeddah, 21461, Saudi Arabia
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - M.A. Alghamdi
- College of Medicine, King Khalid University, Abha, 62529, Saudi Arabia
- Genomics and Personalized Medicine Unit, The Center for Medical and Health Research, King Khalid University, Abha, 62529, Saudi Arabia
| | - I.M. Khalifa
- Clinical Sciences Department, MBBS Program, Fakeeh College for Medical Sciences, Jeddah, 21461, Saudi Arabia
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - W.A. Abd Algaleel
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Faculty of Medicine, Galala University, Suez, Egypt
| |
Collapse
|
36
|
Madreiter-Sokolowski CT, Hiden U, Krstic J, Panzitt K, Wagner M, Enzinger C, Khalil M, Abdellatif M, Malle E, Madl T, Osto E, Schosserer M, Binder CJ, Olschewski A. Targeting organ-specific mitochondrial dysfunction to improve biological aging. Pharmacol Ther 2024; 262:108710. [PMID: 39179117 DOI: 10.1016/j.pharmthera.2024.108710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/09/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
In an aging society, unveiling new anti-aging strategies to prevent and combat aging-related diseases is of utmost importance. Mitochondria are the primary ATP production sites and key regulators of programmed cell death. Consequently, these highly dynamic organelles play a central role in maintaining tissue function, and mitochondrial dysfunction is a pivotal factor in the progressive age-related decline in cellular homeostasis and organ function. The current review examines recent advances in understanding the interplay between mitochondrial dysfunction and organ-specific aging. Thereby, we dissect molecular mechanisms underlying mitochondrial impairment associated with the deterioration of organ function, exploring the role of mitochondrial DNA, reactive oxygen species homeostasis, metabolic activity, damage-associated molecular patterns, biogenesis, turnover, and dynamics. We also highlight emerging therapeutic strategies in preclinical and clinical tests that are supposed to rejuvenate mitochondrial function, such as antioxidants, mitochondrial biogenesis stimulators, and modulators of mitochondrial turnover and dynamics. Furthermore, we discuss potential benefits and challenges associated with the use of these interventions, emphasizing the need for organ-specific approaches given the unique mitochondrial characteristics of different tissues. In conclusion, this review highlights the therapeutic potential of addressing mitochondrial dysfunction to mitigate organ-specific aging, focusing on the skin, liver, lung, brain, skeletal muscle, and lung, as well as on the reproductive, immune, and cardiovascular systems. Based on a comprehensive understanding of the multifaceted roles of mitochondria, innovative therapeutic strategies may be developed and optimized to combat biological aging and promote healthy aging across diverse organ systems.
Collapse
Affiliation(s)
| | - Ursula Hiden
- Department of Obstetrics and Gynecology, Research Unit of Early Life Determinants, Medical University of Graz, Austria
| | - Jelena Krstic
- Division of Cell Biology, Histology and Embryology, Medical University of Graz, BioTechMed-Graz, Austria
| | - Katrin Panzitt
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Austria
| | - Martin Wagner
- Division of Gastroenterology and Hepatology, Medical University of Graz, Austria
| | | | - Michael Khalil
- Department of Neurology, Medical University of Graz, Austria
| | - Mahmoud Abdellatif
- Division of Cardiology, Medical University of Graz, BioTechMed-Graz, Austria
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Medical University of Graz, BioTechMed-Graz, Austria
| | - Tobias Madl
- Division of Medicinal Chemistry, Medical University of Graz, BioTechMed-Graz, Austria
| | - Elena Osto
- Division of Physiology and Pathophysiology, Medical University of Graz
| | - Markus Schosserer
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Andrea Olschewski
- Department of Anesthesiology and Intensive Care Medicine, LBI for Lung Vascular Research, Medical University of Graz, Austria.
| |
Collapse
|
37
|
Li P, Li D, Lu Y, Pan S, Cheng F, Li S, Zhang X, Huo J, Liu D, Liu Z. GSTT1/GSTM1 deficiency aggravated cisplatin-induced acute kidney injury via ROS-triggered ferroptosis. Front Immunol 2024; 15:1457230. [PMID: 39386217 PMCID: PMC11461197 DOI: 10.3389/fimmu.2024.1457230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 08/23/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Cisplatin is a widely used chemotherapeutic agent prescribed to treat solid tumors. However, its clinical application is limited because of cisplatin- induced nephrotoxicity. A known complication of cisplatin is acute kidney injury (AKI). Deletion polymorphisms of GSTM1 and GSTT1, members of the glutathione S-transferase family, are common in humans and are presumed to be associated with various kidney diseases. However, the specific roles and mechanisms of GSTM1 and GSTT1 in cisplatin induced AKI remain unclear. Methods To investigate the roles of GSTM1 and GSTT1 in cisplatin-induced AKI, we generated GSTM1 and GSTT1 knockout mice using CRISPR-Cas9 technology and assessed their kidney function under normal physiological conditions and cisplatin treatment. Using ELISA kits, we measured the levels of oxidative DNA and protein damage, along with MDA, SOD, GSH, and the GSH/GSSG ratio in wild-type and GSTM1/GSTT1 knockout mice following cisplatin treatment. Additionally, oxidative stress levels and the expression of ferroptosis-related proteins in kidney tissues were examined through Western blotting, qPCR, immunohistochemistry, and immunofluorescence techniques. Results Here, we found that GSTT1 and GSTM1 were downregulated in the renal tubular cells of AKI patients and cisplatin-treated mice. Compared with WT mice, Gstm1/Gstt1-DKO mice were phenotypically normal but developed more severe kidney dysfunction and exhibited increased ROS levels and severe ferroptosis after injecting cisplatin. Discussion Our study revealed that GSTM1 and GSTT1 can protect renal tubular cells against cisplatin-induced nephrotoxicity and ferroptosis, and genetic screening for GSTM1 and GSTT1 polymorphisms can help determine a standard cisplatin dose for cancer patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Peipei Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Duopin Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Yanfang Lu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Fei Cheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shen Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaonan Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Jinling Huo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| |
Collapse
|
38
|
Miao J, Zhu H, Wang J, Chen J, Han F, Lin W. Experimental models for preclinical research in kidney disease. Zool Res 2024; 45:1161-1174. [PMID: 39257378 PMCID: PMC11491777 DOI: 10.24272/j.issn.2095-8137.2024.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/04/2024] [Indexed: 09/12/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are significant public health issues associated with a long-term increase in mortality risk, resulting from various etiologies including renal ischemia, sepsis, drug toxicity, and diabetes mellitus. Numerous preclinical models have been developed to deepen our understanding of the pathophysiological mechanisms and therapeutic approaches for kidney diseases. Among these, rodent models have proven to be powerful tools in the discovery of novel therapeutics, while the development of kidney organoids has emerged as a promising advancement in the field. This review provides a comprehensive analysis of the construction methodologies, underlying biological mechanisms, and recent therapeutic developments across different AKI and CKD models. Additionally, this review summarizes the advantages, limitations, and challenges inherent in these preclinical models, thereby contributing robust evidence to support the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Jin Miao
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Huanhuan Zhu
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Junni Wang
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Jianghua Chen
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Fei Han
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China. E-mail:
| | - Weiqiang Lin
- Department of Nephrology, Center for Regeneration and Aging Medicine, Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang 322000, China. E-mail:
| |
Collapse
|
39
|
Mishra S, Shelke V, Gaikwad AB. Acyl-CoA Synthetase Long-Chain Isoenzymes in Kidney Diseases: Mechanistic Insights and Therapeutic Implications. Cell Biochem Funct 2024; 42:e4114. [PMID: 39210707 DOI: 10.1002/cbf.4114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Long-chain acyl-CoA synthetases (ACSLs) are pivotal enzymes in fatty acid metabolism, essential for maintaining cellular homeostasis and energy production. Recent research has uncovered their significant involvement in the pathophysiology of various kidney diseases, including acute kidney injury (AKI), chronic kidney disease (CKD), diabetic kidney disease (DKD), and renal cell carcinoma (RCC). While ACSL1, ACSL3, ACSL4, and ACSL5 have been extensively studied for their roles in processes such as ferroptosis, lipid peroxidation, renal fibrosis, epithelial-mesenchymal transition, and tumor progression, the role of ACSL6 in kidney diseases remain largely unexplored. Notably, these isoenzymes exhibit distinct functions in different kidney diseases. Therefore, to provide a comprehensive understanding of their involvement, this review highlights the molecular pathways influenced by ACSLs and their roles in modulating cell death, inflammation, and fibrosis during kidney disease progression. By examining these mechanisms in detail, this review underscores the potential of ACSLs as biomarkers and therapeutic targets, advocating for further research to elucidate the precise roles of individual ACSL isoenzymes in kidney disease progression. Understanding these mechanisms opens new avenues for developing targeted interventions and improving therapeutic outcomes for patients with kidney diseases.
Collapse
Affiliation(s)
- Swati Mishra
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Vishwadeep Shelke
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| |
Collapse
|
40
|
Yong X, Zhang Y, Tang H, Hu H, Song R, Wu Q. CDKN2A inhibited ferroptosis through activating JAK2/STAT3 pathway to modulate cisplatin resistance in cervical squamous cell carcinoma. Anticancer Drugs 2024; 35:698-708. [PMID: 38748610 DOI: 10.1097/cad.0000000000001620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Cervical squamous cell carcinoma (CESC) is a significant threat to women's health. Resistance to cisplatin (DDP), a common treatment, hinders the therapeutic efficacy. Understanding the molecular basis of DDP resistance in CESC is imperative. Cyclin-dependent kinase inhibitor 2A (CDKN2A) expression was evaluated through quantitative real-time-PCR and western blot in clinical samples from 30 CESC patients and human cervical epithelial cells and CESC cell lines (SiHa, C33A, and Caski). It was also evaluated through bioinformatics analysis in Timer, Ualcan, and GEPIA database. Cell viability was detected by CCK-8. Apoptosis was detected by Calcein AM/PI assay. Lipid reactive oxygen species (ROS), malondialdehyde, glutathione, Fe 2+ , and iron level were detected by kits. Protein level of JAK2, STAT3, p-JAK2, p-STAT3, ACSL4, GPX4, SLC7A11, and FTL were detected by western blot. In CESC, elevated CDKN2A expression was observed. Cisplatin exhibited a dual effect, inhibiting cell proliferation and inducing ferroptosis in CESC. CDKN2A knockdown in a cisplatin-resistant cell line suppressed proliferation and induced ferroptosis. Moreover, CDKN2A was identified as an inhibitor of erastin-induced ferroptosis. Additionally, targeting the JAK2/STAT3 pathway enhanced ferroptosis in cisplatin-resistant cells. CDKN2A could inhibit ferroptosis in CESC through activating JAK2/STAT3 pathway to modulate cisplatin resistance.
Collapse
Affiliation(s)
- Xiang Yong
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei City
- Department of Pathology, Anhui Wanbei Coal-Electricity Group General Hospital
- Department of Tumor Pathology, Suzhou City Key Laboratory of Tumor Pathology
| | - Yanling Zhang
- Department of Oncology, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou City, Anhui Province, China
| | - Heng Tang
- Department of Pathology, Anhui Wanbei Coal-Electricity Group General Hospital
- Department of Tumor Pathology, Suzhou City Key Laboratory of Tumor Pathology
| | - Huaiyuan Hu
- Department of Pathology, Anhui Wanbei Coal-Electricity Group General Hospital
- Department of Tumor Pathology, Suzhou City Key Laboratory of Tumor Pathology
| | - Rui Song
- Department of Pathology, Anhui Wanbei Coal-Electricity Group General Hospital
- Department of Tumor Pathology, Suzhou City Key Laboratory of Tumor Pathology
| | - Qiang Wu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei City
| |
Collapse
|
41
|
Tschuck J, Padmanabhan Nair V, Galhoz A, Zaratiegui C, Tai HM, Ciceri G, Rothenaigner I, Tchieu J, Stockwell BR, Studer L, Cabianca DS, Menden MP, Vincendeau M, Hadian K. Suppression of ferroptosis by vitamin A or radical-trapping antioxidants is essential for neuronal development. Nat Commun 2024; 15:7611. [PMID: 39218970 PMCID: PMC11366759 DOI: 10.1038/s41467-024-51996-1] [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: 08/16/2023] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The development of functional neurons is a complex orchestration of multiple signaling pathways controlling cell proliferation and differentiation. Because the balance of antioxidants is important for neuronal survival and development, we hypothesized that ferroptosis must be suppressed to gain neurons. We find that removal of antioxidants diminishes neuronal development and laminar organization of cortical organoids, which is fully restored when ferroptosis is inhibited by ferrostatin-1 or when neuronal differentiation occurs in the presence of vitamin A. Furthermore, iron-overload-induced developmental growth defects in C. elegans are ameliorated by vitamin E and A. We determine that all-trans retinoic acid activates the Retinoic Acid Receptor, which orchestrates the expression of anti-ferroptotic genes. In contrast, retinal and retinol show radical-trapping antioxidant activity. Together, our study reveals an unexpected function of vitamin A in coordinating the expression of essential cellular gatekeepers of ferroptosis, and demonstrates that suppression of ferroptosis by radical-trapping antioxidants or by vitamin A is required to obtain mature neurons and proper laminar organization in cortical organoids.
Collapse
Affiliation(s)
- Juliane Tschuck
- Research Unit Signaling and Translation, Helmholtz Zentrum München, Neuherberg, Germany
| | - Vidya Padmanabhan Nair
- Endogenous Retrovirus Group, Institute of Virology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ana Galhoz
- Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Biology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Carole Zaratiegui
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Hin-Man Tai
- Endogenous Retrovirus Group, Institute of Virology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gabriele Ciceri
- Developmental Biology and Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ina Rothenaigner
- Research Unit Signaling and Translation, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jason Tchieu
- Developmental Biology and Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- UC Department of Pediatrics, Division of Developmental Biology, Cincinnati Children's Hospital Medical, Cincinnati, OH, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Department of Chemistry, Herbert Irving Comprehensive Cancer Center, Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Lorenz Studer
- Developmental Biology and Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daphne S Cabianca
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Michael P Menden
- Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville Victoria, Australia
| | - Michelle Vincendeau
- Endogenous Retrovirus Group, Institute of Virology, Helmholtz Zentrum München, Neuherberg, Germany.
- Technical University of Munich, Institute of Virology, School of Medicine, Munich, Germany.
| | - Kamyar Hadian
- Research Unit Signaling and Translation, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
42
|
Deng Y, Lu L, Zhu D, Zhang H, Fu Y, Tan Y, Tan X, Guo M, Zhang Y, Yang H, Yang B, Liu T, Chen Y. MafG/MYH9-LCN2 axis promotes liver fibrosis through inhibiting ferroptosis of hepatic stellate cells. Cell Death Differ 2024; 31:1127-1139. [PMID: 38871948 PMCID: PMC11369194 DOI: 10.1038/s41418-024-01322-5] [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: 10/26/2023] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024] Open
Abstract
Hepatic stellate cells (HSCs) secrete extracellular matrix for collagen deposition, contributing to liver fibrosis. Ferroptosis is a novel type of programmed cell death induced by iron overload-dependent lipid peroxidation. Regulation of ferroptosis in hepatic stellate cells (HSCs) may have therapeutic potential for liver fibrosis. Here, we found that Maf bZIP transcription factor G (MafG) was upregulated in human and murine liver fibrosis. Interestingly, MafG knockdown increased HSCs ferroptosis, while MafG overexpression conferred resistance of HSCs to ferroptosis. Mechanistically, MafG physically interacted with non-muscle myosin heavy chain IIa (MYH9) to transcriptionally activate lipocalin 2 (LCN2) expression, a known suppressor for ferroptosis. Site-directed mutations of MARE motif blocked the binding of MafG to LCN2 promoter. Re-expression of LCN2 in MafG knockdown HSCs restored resistance to ferroptosis. In bile duct ligation (BDL)-induced mice model, we found that treatment with erastin alleviated murine liver fibrosis by inducing HSC ferroptosis. HSC-specific knowdown MafG based on adeno-associated virus 6 (AAV-6) improved erastin-induced HSC ferroptosis and alleviation of liver fibrosis. Taken together, MafG inhibited HSCs ferroptosis to promote liver fibrosis through transcriptionally activating LCN2 expression. These results suggest that MafG/MYH9-LCN2 signaling pathway could be a novel targets for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yalan Deng
- Department of Ultrasonic Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Liqing Lu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Dandan Zhu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huajun Zhang
- Department of Ultrasonic Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ying Fu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yuying Tan
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xuemei Tan
- Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ming Guo
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ye Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Heping Yang
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Bing Yang
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ting Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
43
|
Guo RZ, Li J, Pan SK, Hu MY, Lv LX, Feng Q, Qiao YJ, Duan JY, Liu DW, Liu ZS. Liquiritigenin, an Active Ingredient of Liquorice, Alleviates Acute Kidney Injury by VKORC1-Mediated Ferroptosis Inhibition. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1507-1526. [PMID: 39192677 DOI: 10.1142/s0192415x24500599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Acute kidney injury (AKI) is a major public health problem worldwide that still lacks effective treatments. Recent studies have suggested that ferroptosis is a key mediator of AKI due to its activation of lipid peroxidation. Therefore, we hypothesized that antiferroptosis agents might be a novel potential therapeutic strategy for AKI. Herein, we demonstrated that liquiritigenin (LG), an active ingredient of liquorice, improves renal function by inhibiting vitamin K epoxide reductase complex subunit 1 (VKORC1)-mediated ferroptosis, both in vivo and in vitro. In a folic acid-induced murine AKI model, after a single pre-treatment intravenous injection, LG markedly alleviated the loss of renal function through suppressing ferroptosis induced by iron accumulation. LG prevented mitochondrial morphological changes and upregulated glutathione and glutathione peroxidase 4 levels, while downregulating malonaldehyde and divalent iron levels. An in vitro RNA-sequence analysis suggested that the protective role of LG may involve upregulation of VKORC1. Moreover, knockdown of VKORC1 diminished the renal protective and antiferroptosis roles of LG. Collectively, our findings demonstrated that LG protected against AKI by inhibiting VKORC1-mediated ferroptosis. This suggests that inhibiting ferroptosis might be a novel therapeutic approach in the future.
Collapse
Affiliation(s)
- Run-Zhi Guo
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Jia Li
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Shao-Kang Pan
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Ming-Yang Hu
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Lin-Xiao Lv
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Qi Feng
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Ying-Jin Qiao
- Blood Purification Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Jia-Yu Duan
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Dong-Wei Liu
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Zhang-Suo Liu
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| |
Collapse
|
44
|
Liu R, Wang J, Liu Y, Gao Y, Yang R. Regulation of gut microbiota on immune cell ferroptosis: A novel insight for immunotherapy against tumor. Cancer Lett 2024; 598:217115. [PMID: 39025428 DOI: 10.1016/j.canlet.2024.217115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024]
Abstract
Gut microbiota contributes to the homeostasis of immune system and is related to various diseases such as tumorigenesis. Ferroptosis, a new type of cell death, is also involved in the disease pathogenesis. Recent studies have found the correlations of gut microbiota mediated ferroptosis and immune cell death. Gut microbiota derived immunosuppressive metabolites, which can promote differentiation and function of immune cells, tend to inhibit ferroptosis through their receptors, whereas inflammatory metabolites from gut microbiota also affect the differentiation and function of immune cells and their ferroptosis. Thus, it is possible for gut microbiota to regulate immune cell ferroptosis. Indeed, gut microbiota metabolite receptor aryl hydrocarbon receptor (AhR) can affect ferroptosis of intestinal intraepithelial lymphocytes, leading to disease pathogenesis. Since immune cell ferroptosis in tumor microenvironment (TME) affects the occurrence and development of tumor, the modulation of gut microbiota in these cell ferroptosis might influence on the tumorigenesis, and also immunotherapy against tumors. Here we will summarize the recent advance of ferroptosis mediated by gut microbiota metabolites, which potentially acts as regulator(s) on immune cells in TME for therapy against tumor.
Collapse
Affiliation(s)
- Ruobing Liu
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Yuqing Liu
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Yunhuan Gao
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin 300071, China.
| |
Collapse
|
45
|
You L. Dihydromyricetin Inhibits Ferroptosis to Attenuate Cisplatin-Induced Muscle Atrophy. Physiol Res 2024; 73:405-413. [PMID: 39027957 PMCID: PMC11299785 DOI: 10.33549/physiolres.935317] [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: 01/03/2024] [Accepted: 05/14/2024] [Indexed: 07/27/2024] Open
Abstract
Cisplatin is a widely used chemotherapy drug for the treatment of various cancers. However, although cisplatin is effective in targeting cancer cells, it has severe side effects including skeletal muscle atrophy. In this study, we aimed to characterize the role of Dihydromyricetin in cisplatin-induced muscle atrophy in mice. 5-week-old male C57BL/6 mice were treated with Dihydromyricetin for 14 days orally followed by in intraperitoneally cisplatin administration for 6 days. Gastrocnemius muscles were isolated for the following experiments. Antioxidative stress were determined by peroxidative product malondialdehyde (MDA) and antioxidants superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities. Quadriceps muscle mass and grip strength were significantly restored by Dihydromyricetin in a dose-dependent manner. Moreover, muscle fibers were improved in Dihydromyricetin treated group. Excessive skeletal muscle E3 ubiquitin-protein ligases in cisplatin group were significantly repressed by Dihydromyricetin treatment. Dihydromyricetin significantly reduced oxidative stress induced by cisplatin by decreasing MDA level and restored SOD and GPx activities. In addition, ferroptosis was significantly reduced by Dihydromyricetin characterized by reduced iron level and ferritin heavy chain 1 and improved Gpx4 level. The present study demonstrated that Dihydromyricetin attenuated cisplatin-induced muscle atrophy by reducing skeletal muscle E3 ubiquitin-protein ligases, oxidative stress, and ferroptosis.
Collapse
Affiliation(s)
- L You
- The Second Affiliated Hospital, Department of Clinical Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
46
|
Li Y, Zhang K, Feng Y, Wu L, Jia Y, Zhao R. Alisma Orientalis Extract Ameliorates Hepatic Iron Deregulation in MAFLD Mice via FXR-Mediated Gene Repression. Nutrients 2024; 16:2272. [PMID: 39064715 PMCID: PMC11279993 DOI: 10.3390/nu16142272] [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: 05/26/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Iron is a vital trace element for our bodies and its imbalance can lead to various diseases. The progression of metabolic-associated fatty liver disease (MAFLD) is often accompanied by disturbances in iron metabolism. Alisma orientale extract (AOE) has been reported to alleviate MAFLD. However, research on its specific lipid metabolism targets and its potential impact on iron metabolism during the progression of MAFLD remains limited. To establish a model of MAFLD, mice were fed either a standard diet (CON) or a high-fat diet (HFD) for 9 weeks. The mice nourished on the HFD were then randomly assigned to the HF group and the HFA group, with the HFA group receiving AOE by gavage on a daily basis for 13 weeks. Supplementation with AOE remarkably reduced overabundant lipid accumulation in the liver and restored the iron content of the liver. AOE partially but significantly reversed dysregulated lipid metabolizing genes (SCD1, PPAR γ, and CD36) and iron metabolism genes (TFR1, FPN, and HAMP) induced by HFD. Chromatin immunoprecipitation assays indicated that the reduced enrichment of FXR on the promoters of SCD1 and FPN genes induced by HFD was significantly reversed by AOE. These findings suggest that AOE may alleviate HFD-induced disturbances in liver lipid and iron metabolism through FXR-mediated gene repression.
Collapse
Affiliation(s)
- Yanlin Li
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (K.Z.); (Y.F.); (L.W.); (Y.J.)
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ke Zhang
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (K.Z.); (Y.F.); (L.W.); (Y.J.)
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yue Feng
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (K.Z.); (Y.F.); (L.W.); (Y.J.)
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Lei Wu
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (K.Z.); (Y.F.); (L.W.); (Y.J.)
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yimin Jia
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (K.Z.); (Y.F.); (L.W.); (Y.J.)
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (K.Z.); (Y.F.); (L.W.); (Y.J.)
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- National Key Laboratory of Meat Quality Control and Cultured Meat Development, Nanjing 210095, China
| |
Collapse
|
47
|
Li J, Fu C, Feng B, Liu Q, Gu J, Khan MN, Sun L, Wu H, Wu H. Polyacrylic Acid-Coated Selenium-Doped Carbon Dots Inhibit Ferroptosis to Alleviate Chemotherapy-Associated Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400527. [PMID: 38689508 PMCID: PMC11267338 DOI: 10.1002/advs.202400527] [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: 01/15/2024] [Revised: 03/26/2024] [Indexed: 05/02/2024]
Abstract
Cisplatin-associated acute kidney injury (AKI) is a severe clinical syndrome that significantly restricts the chemotherapeutic application of cisplatin in cancer patients. Ferroptosis, a newly characterized programmed cell death driven by the lethal accumulation of lipid peroxidation, is widely reported to be involved in the pathogenesis of cisplatin-associated AKI. Targeted inhibition of ferroptosis holds great promise for developing novel therapeutics to alleviate AKI. Unfortunately, current ferroptosis inhibitors possess low bioavailability or perform non-specific accumulation in the body, making them inefficient in alleviating cisplatin-associated AKI or inadvertently reducing the anti-tumor efficacy of cisplatin, thus not suitable for clinical application. In this study, a novel selenium nanomaterial, polyacrylic acid-coated selenium-doped carbon dots (SeCD), is rationally developed. SeCD exhibits high biocompatibility and specifically accumulates in the kidney. Administration of SeCD effectively scavenges broad-spectrum reactive oxygen species and significantly facilitates GPX4 expression by releasing selenium, resulting in strong mitigation of ferroptosis in renal tubular epithelial cells and substantial alleviation of cisplatin-associated AKI, without compromising the chemotherapeutic efficacy of cisplatin. This study highlights a novel and promising therapeutic approach for the clinical prevention of AKI in cancer patients undergoing cisplatin chemotherapy.
Collapse
Affiliation(s)
- Jiahuan Li
- State Key Laboratory of Agricultural MicrobiologyCollege of Animal Science & Technology and College of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryWuhan430070China
| | - Chengcheng Fu
- Hubei Hongshan LaboratoryWuhan430070China
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze RiverCollege of Plant Science & TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureGenome Analysis Laboratory of the Ministry of AgricultureAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518120China
| | - Baoli Feng
- State Key Laboratory of Agricultural MicrobiologyCollege of Animal Science & Technology and College of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryWuhan430070China
| | - Qingquan Liu
- Department of NephrologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Jiangjiang Gu
- College of ChemistryHuazhong Agricultural UniversityWuhan430070China
| | - Mohammad Nauman Khan
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication)Hainan UniversitySanya572000China
| | - Lvhui Sun
- State Key Laboratory of Agricultural MicrobiologyCollege of Animal Science & Technology and College of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryWuhan430070China
| | - Honghong Wu
- Hubei Hongshan LaboratoryWuhan430070China
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze RiverCollege of Plant Science & TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureGenome Analysis Laboratory of the Ministry of AgricultureAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518120China
| | - Hao Wu
- State Key Laboratory of Agricultural MicrobiologyCollege of Animal Science & Technology and College of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryWuhan430070China
| |
Collapse
|
48
|
Du K, Wang L, Jun JH, Dutta RK, Maeso-Díaz R, Oh SH, Ko DC, Diehl AM. Aging promotes metabolic dysfunction-associated steatotic liver disease by inducing ferroptotic stress. NATURE AGING 2024; 4:949-968. [PMID: 38918603 DOI: 10.1038/s43587-024-00652-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/17/2024] [Indexed: 06/27/2024]
Abstract
Susceptibility to the biological consequences of aging varies among organs and individuals. We analyzed hepatocyte transcriptomes of healthy young and aged male mice to generate an aging hepatocyte gene signature, used it to deconvolute transcriptomic data from humans and mice with metabolic dysfunction-associated liver disease, validated findings with functional studies in mice and applied the signature to transcriptomic data from other organs to determine whether aging-sensitive degenerative mechanisms are conserved. We discovered that the signature enriches in diseased livers in parallel with degeneration. It is also enriched in failing human hearts, diseased kidneys and pancreatic islets from individuals with diabetes. The signature includes genes that control ferroptosis. Aged mice develop more hepatocyte ferroptosis and liver degeneration than young mice when fed diets that induce metabolic stress. Inhibiting ferroptosis shifts the liver transcriptome of old mice toward that of young mice and reverses aging-exacerbated liver damage, identifying ferroptosis as a tractable, conserved mechanism for aging-related tissue degeneration.
Collapse
Affiliation(s)
- Kuo Du
- Department of Medicine, Duke University, Durham, NC, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Ji Hye Jun
- Department of Medicine, Duke University, Durham, NC, USA
| | - Rajesh K Dutta
- Department of Medicine, Duke University, Durham, NC, USA
| | | | - Seh Hoon Oh
- Department of Medicine, Duke University, Durham, NC, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
49
|
Zhang J, Che T, Wang L, Sun W, Zhao J, Chen J, Liu Y, Pu Q, Zhang Y, Li J, Li Z, Zhu Z, Fu Q, Wang X, Yuan J. Proteomics coupled transcriptomics reveals Slc34a1 and Slc34a3 downregulation as potential features of nephrotoxin-induced acute kidney injury. J Proteomics 2024; 302:105203. [PMID: 38782357 DOI: 10.1016/j.jprot.2024.105203] [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: 08/07/2023] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Acute kidney injury (AKI) stands as a prevalent and economically burdensome condition worldwide, yet its complex molecular mechanisms remain incompletely understood. To address this gap, our study employs a multifaceted approach, combining mass spectrometry and RNA sequencing technologies, to elucidate the intricate molecular landscape underlying nephrotoxin-induced AKI in mice by cisplatin- and LPS-induced. By examining the protein and RNA expression profiles, we aimed to uncover novel insights into the pathogenesis of AKI and identify potential diagnostic and therapeutic targets. Our results demonstrate significant down-regulation of Slc34a1 and Slc34a3, shedding light on their crucial roles in AKI pathology and highlighting their promise as actionable targets for diagnosis and treatment. This comprehensive analysis not only enhances our understanding of AKI pathophysiology but also offers valuable avenues for the development of targeted interventions to mitigate its clinical impact. SIGNIFICANCE: Nephrotoxicity acute kidney injury (AKI) is a common clinical condition whose pathogenesis is the process by which some drugs, chemicals or other factors cause damage to the kidneys, resulting in impaired kidney function. Although it has been proved that different nephrotoxic substances can affect the kidney through different pathways, whether they have a commonality has not been registered. Here, we combined transcriptomics and proteomics to study the molecular mechanism of LPS and cisplatin-induced nephrotoxic acute kidney injury finding that the down-regulation of Slc34a1 and Slc34a3 may be a critical link in nephrotoxic acute kidney injury, which can be used as a marker for its early diagnosis.
Collapse
Affiliation(s)
- Junying Zhang
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China; College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Tiantian Che
- Chongqing Nanan District Center for Diseases Control and Prevention, Chongqing 401336, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Wei Sun
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jing Zhao
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jiajia Chen
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Yang Liu
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Qi Pu
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Yu Zhang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jiani Li
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhangfu Li
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China; Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, Shenzhen 518036, China
| | - Zhaojing Zhu
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| | - Qihuan Fu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China.
| | - Xiaoyang Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China..
| | - Jiangbei Yuan
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China; Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, Shenzhen 518036, China.; Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Department of Infectious Diseases, Affiliated Banan Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
50
|
Guo J, Le Y, Yuan A, Liu J, Chen H, Qiu J, Wang C, Dou X, Yuan X, Lu D. Astragaloside IV ameliorates cisplatin-induced liver injury by modulating ferroptosis-dependent pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118080. [PMID: 38521426 DOI: 10.1016/j.jep.2024.118080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/06/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The use of antineoplastic drugs, such as cisplatin, in clinical practice can cause adverse effects in patients, such as liver injury, which limits their long-term use. Therefore, there is an urgent need to develop alternative therapeutic strategies or drugs to minimize cisplatin-induced liver injury. Huangqi, the root of Astragalus membranaceus, is extensively used in traditional Chinese medicine (TCM) and has been employed in treating diverse liver injuries. Astragalus membranaceus contains several bioactive constituents, including triterpenoid saponins, one of which, astragaloside IV (ASIV), has been reported to have anti-inflammatory and antioxidant stress properties. However, its potential in ameliorating cisplatin-induced liver injury has not been explored. AIM OF THE STUDY The objective of this study was to examine the mechanism by which ASIV protects against cisplatin-induced liver injury. MATERIALS AND METHODS This study established a model of cisplatin-induced liver injury in mice, followed by treatment with various doses of astragaloside IV (40 mg/kg, 80 mg/kg). In addition, a model of hepatocyte ferroptosis in AML-12 cells was established using RSL3. The mechanism of action of astragaloside IV was investigated using a range of methods, including Western blot assay, qPCR, immunofluorescence, histochemistry, molecular docking, and high-content imaging system. RESULTS The findings suggested a significant improvement in hepatic injury, inflammation and oxidative stress phenotypes with the administration of ASIV. Furthermore, network pharmacological analyses provided evidence that a major pathway for ASIV to attenuate cisplatin-induced hepatic injury entailed the cell death cascade pathway. It was observed that ASIV effectively inhibited ferroptosis both in vivo and in vitro. Subsequent experimental outcomes provided further validation of ASIV's ability to hinder ferroptosis through the inhibition of PPARα/FSP1 signaling pathway. The current findings suggest that ASIV could function as a promising phytotherapy composition to alleviate cisplatin-induced liver injury. CONCLUSIONS The current findings suggest that astragaloside IV could function as a promising phytotherapy composition to alleviate cisplatin-induced liver injury.
Collapse
Affiliation(s)
- Jianan Guo
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Yifei Le
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Aini Yuan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Jing Liu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Hang Chen
- Department of Medical Research Center, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China.
| | - Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Cui Wang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Xingyu Yuan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Dezhao Lu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|