1
|
Lu Q, Shao N, Fang Z, Ouyang Z, Shen Y, Yang R, Liu H, Cai B, Wei T. The anti-Alzheimer's disease effects of ganoderic acid A by inhibiting ferroptosis-lipid peroxidation via activation of the NRF2/SLC7A11/GPX4 signaling pathway. Chem Biol Interact 2025; 412:111459. [PMID: 40054827 DOI: 10.1016/j.cbi.2025.111459] [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: 01/03/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system, characterized by a gradual decline in cognitive and memory abilities, social disorders, and behavioral abnormalities. Ferroptosis, an iron-dependent type of programmed cell death, is closely associated with the pathogenesis of AD. Ferroptosis is characterized by the accumulation of iron within cells, leading to increased oxidative stress, and ultimately lipid peroxidation and cell death. Ganoderic acid A (GAA), one of the major pharmacologically active components in Ganoderma lucidum, exhibits an excellent neuroprotective effect against AD. However, it is unclear whether GAA improves the symptoms of AD by inhibiting ferroptosis. This study investigated the anti-AD effects of GAA through both in vivo and in vitro experiments, and determined its molecular mechanism from the perspective of ferroptosis modulation. The results showed that GAA administration attenuated hippocampal neuronal loss, improved mitochondrial ultrastructure, and enhanced the memory and learning ability of the AD mice. In vitro assays suggested that GAA effectively protected HT22 AD cells against ferroptosis-related morphological damage, enhanced their antioxidant capacity, maintained their iron metabolism, and reduced mitochondrial dysfunction. Moreover, the immunofluorescence and western blotting results showed that the levels of NFE2 like bZIP transcription factor 2 (NRF2), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11) both in the hippocampus of APP/PS1 mice and amyloid beta (Aβ)25-35-induced HT22 AD cells were markedly enhanced after GAA administration. In summary, these results revealed that GAA improves AD by activating on the NRF2/SLC7A11/GPX4 axis to inhibit ferroptosis-lipid peroxidation.
Collapse
Affiliation(s)
- Qingyang Lu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Nan Shao
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ziyi Fang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Zhaorong Ouyang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yiran Shen
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ruiling Yang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Houli Liu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Biao Cai
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Tao Wei
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
2
|
Coradduzza D, Sanna A, Di Lorenzo B, Congiargiu A, Marra S, Cossu M, Tedde A, De Miglio MR, Zinellu A, Mangoni AA, Cogoni AA, Madonia M, Carru C, Medici S. Associations between plasma and urinary heavy metal concentrations and the risk of prostate cancer. Sci Rep 2025; 15:14274. [PMID: 40274938 PMCID: PMC12022027 DOI: 10.1038/s41598-025-97682-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Prostate cancer is a leading cause of morbidity and mortality worldwide, with incidence rates projected to double between 2020 and 2040. This growing health challenge highlights the need for improved diagnostic strategies and risk assessment tools to better understand disease etiology and progression. Among environmental factors, heavy metals have been implicated in inflammation and carcinogenesis, yet their specific role in prostate disease remains insufficiently explored. This novel study analyses the relationship between heavy metal concentrations in plasma and urine of patients with benign prostatic hyperplasia, precancerous lesions and prostate cancer. In addition to evaluating key clinical parameters, including age, total PSA levels, hemoglobin concentrations and monocyte/lymphocyte ratio, it aims to determine whether specific heavy metals contribute to the progression of prostate disease and whether they can serve as potential biomarkers for early diagnosis. Our findings reveal significant differences in vanadium and antimony concentrations in plasma, suggesting a potential role in prostate disease pathophysiology. Notably, lower plasma antimony concentrations are associated with an increased risk of PC, while plasma vanadium concentrations are significantly higher in the PL group. Regression analysis further supports the association between heavy metal concentrations and the risk of PL and PC, highlighting the potential of vanadium and copper as biomarkers or therapeutic targets for prostate health. The study also explores the impact of lead exposure on prostate cancer risk, revealing a significant association between urine lead concentration and PC. These findings underscore the complex interaction between heavy metal concentrations and prostate disease risk, emphasizing the need for further research to elucidate underlying mechanisms and explore therapeutic interventions.
Collapse
Affiliation(s)
| | - Andrea Sanna
- SC Chimica Istituto Zooprofilattico Sperimentale della Sardegna, Via Duca degli Abruzzi, 8, 07100, Sassari, Italy
| | - Biagio Di Lorenzo
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | | | - Sonia Marra
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
- Unit of Urology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy
| | - Maurizio Cossu
- SC Chimica Istituto Zooprofilattico Sperimentale della Sardegna, Via Duca degli Abruzzi, 8, 07100, Sassari, Italy
| | - Alessandro Tedde
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
- Unit of Urology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy
| | | | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, Flinders University, Adelaide, Australia
| | - Alessio Aligio Cogoni
- Medical Oncology Department, Azienda Ospedaliero-Universitaria di Sassari, Sassari, Italy
| | - Massimo Madonia
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
- Unit of Urology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- Medical Oncology Department, Azienda Ospedaliero-Universitaria di Sassari, Sassari, Italy
| | - Serenella Medici
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, 07100, Sassari, Italy.
| |
Collapse
|
3
|
Rao Y, Pan Q, Liu S, Yao S, Li L, Yan J, Chen L, Xu L, Yan H, Ma A, Wang F, Mao X, Wang Z, Zhang J, Guo J, Sun Z. Tissue inhibitor of metalloproteinase 1 promotes ferroptosis and suppresses prostate cancer metastasis. J Biol Chem 2025; 301:108473. [PMID: 40185230 DOI: 10.1016/j.jbc.2025.108473] [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: 01/13/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
Tissue inhibitor of metalloproteinase 1 (TIMP1) has been implicated in prostate cancer (PCa) metastasis. In this study, PC-3M-2B4 cells with TIMP1 knockdown (PC-3M-2B4-shTIMP1) or overexpression (PC-3M-2B4-TIMP1) were generated, and an inverse correlation was found between TIMP1 expression and cell migration and invasion, which was confirmed in vitro and in vivo. Differential TIMP1 expression was accompanied by variations in the expression of the ferroptosis-related proteins, glutathione peroxidase 4 (GPX4), transferrin receptor, transferrin, glutamine cysteine ligase catalytic subunit, and glutamine cysteine ligase modifier subunit. In comparison with TIMP1-overexpressing cells, TIMP1-knockdown cells demonstrated a 12.3% decrease in Fe2+ concentration after erastin treatment, a 37.8% reduction in malondialdehyde levels, an 113.7% increase in GPX4 expression, and a 78.9% rise in the GSH-GSSG ratio. Our findings indicate that TIMP1 overexpression promotes ferroptosis by modulating critical markers, such as GPX4 and transferrin receptor, thereby significantly reducing metastatic potential in PCa cells. Our results highlight the role of TIMP1 in regulating ferroptosis pathways, which are crucial for tumor progression, and exposes a potential therapeutic target for PCa management.
Collapse
Affiliation(s)
- Yuliang Rao
- School of Pharmacy, School of Basic Medical Sciences, Fudan University, Shanghai, China; National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Qi Pan
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Siyu Liu
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Shunheng Yao
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Lei Li
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Jianyan Yan
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Lifen Chen
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Li Xu
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Han Yan
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Aicui Ma
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Fen Wang
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Xiaoyan Mao
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Zhonghui Wang
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Junfang Zhang
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Jun Guo
- National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China.
| | - Zuyue Sun
- School of Pharmacy, School of Basic Medical Sciences, Fudan University, Shanghai, China; National Evaluation Centre for the Toxicology of Fertility Regulating Drug, Shanghai Institute for Bio-medical and Pharmaceutical Technologies (SIBPT), Shanghai, China; NHC Key Laboratory of Reproduction Regulation, Shanghai, China; Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai, China.
| |
Collapse
|
4
|
Xu J, Bai X, Dong K, Du Q, Ma P, Zhang Z, Yang J. GluOC Induced SLC7A11 and SLC38A1 to Activate Redox Processes and Resist Ferroptosis in TNBC. Cancers (Basel) 2025; 17:739. [PMID: 40075587 PMCID: PMC11899354 DOI: 10.3390/cancers17050739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Ferroptosis, a type of programmed cell death, is mainly associated with disruptions in iron metabolism, imbalances in the amino acid antioxidant system, and the build-up of lipid peroxides. Triple-negative breast cancer (TNBC) has a dismal prognosis. Since activating ferroptosis can suppress breast cancer cell proliferation, it holds promise as a novel therapeutic target for breast cancer patients. Thus, the objective of this study was to clarify the mechanism of ferroptosis in TNBC, aiming to find new treatment strategies for TNBC patients. METHODS We screened out the differential genes related to ferroptosis in TNBC after GluOC treatment based on the whole-genome sequencing results. At the cellular level, we conducted explorations using techniques such as quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, fluorescence staining, and siRNA transfection. Moreover, we further verified the role of GluOC in inhibiting ferroptosis in TNBC through in vivo experiments using nude mice. RESULTS The results showed that GluOC enhanced glutathione expression levels by inducing SLC7A11 accumulation via the specific signaling pathway. Additionally, GluOC increased ATP production and tricarboxylic acid flux resistance to ferroptosis through SLC38A1. Overall, GluOC coordinately regulated SLC7A11 and SLC38A1 to inhibit ferroptosis in TNBC. CONCLUSIONS This study elucidated the mechanism of GluOC in inhibiting ferroptosis in TNBC. The findings not only provided new insights into ferroptosis but also potentially offered new concepts for the development of future anticancer therapies, which may contribute to improving the treatment of TNBC patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jianhong Yang
- Medical School, University of Chinese Academy of Sciences, No. 1, Yanqi Lake East Road, Huairou District, Beijing 101408, China; (J.X.); (X.B.); (K.D.); (Q.D.); (P.M.); (Z.Z.)
| |
Collapse
|
5
|
Yang H, Zhang X, Jia Z, Wang H, Wu J, Wei X, Huang Y, Yan W, Lin Y. Targeting ferroptosis in prostate cancer management: molecular mechanisms, multidisciplinary strategies and translational perspectives. J Transl Med 2025; 23:166. [PMID: 39920771 PMCID: PMC11806579 DOI: 10.1186/s12967-025-06180-4] [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: 12/06/2024] [Accepted: 01/25/2025] [Indexed: 02/09/2025] Open
Abstract
Prostate cancer (PCa) is a kind of malignant solid tumor commonly observed among males worldwide. The dilemma of increasing incidence with therapeutic resistance has become the leading issue in PCa clinical management. Ferroptosis is a new form of regulatory cell death caused by iron-dependent lipid peroxidation, which has a dual role in PCa evolution and treatment due to the multi-omics cascade of interactions among pathways and environmental stimuli. Hence deciphering the role of ferroptosis in carcinogenesis would provide novel insights and strategies for precision medicine and personalized healthcare against PCa. In this study, the mechanisms of ferroptosis during cancer development were summarized both at the molecular and tumor microenvironment level. Then literature-reported ferroptosis-related signatures in PCa, e.g., genes, non-coding RNAs, metabolites, natural products and drug components, were manually collected and functionally compared as drivers/inducers, suppressors/inhibitors, and biomarkers according to their regulatory patterns in PCa ferroptosis and pathogenesis. The state-of-the-art techniques for ferroptosis-related data integration, knowledge identification, and translational application to PCa theranostics were discussed from a combinative perspective of artificial intelligence-powered modelling and advanced material-oriented therapeutic scheme design. The prospects and challenges in ferroptosis-based PCa researches were finally highlighted to light up future wisdoms for the flourishing of current findings from bench to bedside.
Collapse
Affiliation(s)
- Hubo Yang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Xuefeng Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Zongming Jia
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - He Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Jixiang Wu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Xuedong Wei
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
| | - Wenying Yan
- Suzhou Key Lab of Multi-modal Data Fusion and Intelligent Healthcare, Suzhou, 215104, China.
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Yuxin Lin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Suzhou Key Lab of Multi-modal Data Fusion and Intelligent Healthcare, Suzhou, 215104, China.
| |
Collapse
|
6
|
Gong FH, Liu L, Wang X, Xiang Q, Yi X, Jiang DS. Ferroptosis induced by environmental pollutants and its health implications. Cell Death Discov 2025; 11:20. [PMID: 39856053 PMCID: PMC11759704 DOI: 10.1038/s41420-025-02305-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/19/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Environmental pollution represents a significant public health concern, with the potential health risks associated with environmental pollutants receiving considerable attention over an extended period. In recent years, a substantial body of research has been dedicated to this topic. Since the discovery of ferroptosis, an iron-dependent programmed cell death typically characterized by lipid peroxidation, in 2012, there have been significant advances in the study of its role and mechanism in various diseases. A growing number of recent studies have also demonstrated the involvement of ferroptosis in the damage caused to the organism by environmental pollutants, and the molecular mechanisms involved have been partially elucidated. The targeting of ferroptosis has been demonstrated to be an effective means of ameliorating the health damage caused by PM2.5, organic and inorganic pollutants, and ionizing radiation. This review begins by providing a summary of the most recent and important advances in ferroptosis. It then proceeds to offer a critical analysis of the health effects and molecular mechanisms of ferroptosis induced by various environmental pollutants. Furthermore, as is the case with all rapidly evolving research areas, there are numerous unanswered questions and challenges pertaining to environmental pollutant-induced ferroptosis, which we discuss in this review in an attempt to provide some directions and clues for future research in this field.
Collapse
Affiliation(s)
- Fu-Han Gong
- Department of Cardiology, Tongren People's Hospital, Tongren, Guizhou, China
| | - Liyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuesheng Wang
- Department of Cardiology, Tongren People's Hospital, Tongren, Guizhou, China
| | - Qi Xiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
7
|
Shao W, Liu F, Zhu L, Qian W, Meng Q, Zhang A, Jin S, Lu J, Yan SG. Ferroportin inhibits the proliferation and migration of fibroblast-like synoviocytes in rheumatoid arthritis via regulating ROS/PI3K/AKT signaling pathway. Eur J Pharmacol 2025; 987:177205. [PMID: 39672225 DOI: 10.1016/j.ejphar.2024.177205] [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: 08/13/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/15/2024]
Abstract
The aberrant proliferation of fibroblast-like synoviocytes (FLS) significantly contributes to excessive synovial hyperplasia and joint deformity in rheumatoid arthritis (RA). It has been observed that the membrane iron transporter protein, ferroportin (FPN), is commonly downregulated in tumor cells, while its overexpression can inhibit tumor cell proliferation. However, limited studies have investigated the role of iron in the pathogenesis of RA. In this study, we examined the functional relevance of FPN in RA. The expression of FPN in RA tissue specimens and primary cells was assessed using western blotting and RT-PCR. An adjuvant-induced arthritis (AIA) rat model was established to further validate the expression level of FPN. Phenotypic analysis of FLS cell proliferation was performed via CCK-8, clonogenic formation, and cell scratch assays. The involvement of membrane iron transporter proteins was analyzed through RNAseq and reactive oxygen species (ROS) detection. The results demonstrated decreased expression of FPN in the synovial tissue of RA patients compared to the normal group. Overexpression of FPN can inhibit RA-FLS proliferation and migration by suppressing the PI3K/AKT pathway, and this effect is associated with the elevation of ROS levels. Our findings suggest that the downregulation of FPN may contribute to the pathogenesis of RA, indicating a potential role of iron dysregulation in this disease, and FPN regulates the proliferation and migration of FLS by promoting the levels of ROS in FLS as well as suppressing the PI3K/AKT signaling pathway. These results suggest that FPN could be a potential target for alleviating joint damage in RA.
Collapse
Affiliation(s)
- Weiting Shao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China; School of Life and Science, Anhui Medical University, Ministry of Education, Hefei, China
| | - Fanxiao Liu
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lin Zhu
- School of Life and Science, Anhui Medical University, Ministry of Education, Hefei, China
| | - Weiyi Qian
- School of Life and Science, Anhui Medical University, Ministry of Education, Hefei, China
| | - Qianqian Meng
- School of Life and Science, Anhui Medical University, Ministry of Education, Hefei, China
| | - Afei Zhang
- School of Life and Science, Anhui Medical University, Ministry of Education, Hefei, China
| | - Shuai Jin
- School of Life and Science, Anhui Medical University, Ministry of Education, Hefei, China
| | - Jingtao Lu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China; School of Life and Science, Anhui Medical University, Ministry of Education, Hefei, China.
| | - Shuang G Yan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
8
|
Wang A, Yang J, Deng J, Wang K, Chen G, Lin D. Kaempferol promotes flap survival by inhibiting ferroptosis and inflammation through network pharmacology and in vivo experiments. Wound Repair Regen 2025; 33:e13250. [PMID: 39719508 DOI: 10.1111/wrr.13250] [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: 10/05/2024] [Revised: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 12/26/2024]
Abstract
Skin flap transplantation is a primary method for wound repair; however, postoperative skin flap necrosis remains a significant challenge. Kaempferol, a flavonol abundant in various foods, exhibits diverse pharmacological effects. This study investigated the potential targets of kaempferol for treating skin flap ischemia-reperfusion (I/R) injury through network pharmacology and molecular docking, followed by in vivo validation. Using SwissTargetPredict, PubChem, SymMap V2, and GeneCards databases, 174 potential target proteins of kaempferol were identified. KEGG and GO enrichment analyses, performed using R software, indicated that kaempferol promotes skin flap survival by modulating ferroptosis, TNF-α, and NF-κB signalling pathways. Molecular docking demonstrated stable binding between kaempferol and key proteins, including SIRT1 and NRF2. In vivo, a McFarlane skin flap model was established in Sprague-Dawley rats. Kaempferol treatment improved flap survival, enhanced perfusion areas and distal arteriole visualisation, and increased blood flow in the flap. Furthermore, kaempferol reduced neutrophil infiltration, alleviated oxidative stress, improved mitochondrial morphology and function, and inhibited the release of proinflammatory cytokines. Western blot and immunofluorescence analyses confirmed that kaempferol inhibited ferroptosis and inflammation while promoting flap survival. Mechanistically, kaempferol was found to activate SIRT1-mediated HMGB1/TLR4/NF-κB and NRF2/SLC7A11/GPX4 pathways, thereby promoting skin flap survival and mitigating I/R injury.
Collapse
Affiliation(s)
- An Wang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jialong Yang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiapeng Deng
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Kaitao Wang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Guodong Chen
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Dingsheng Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
9
|
Zhang H, Tang M, Liu Q, Wu D, Sun B, Dong J, Guan L, Luo J, Zeng M. PAT exposure caused human hepatocytes apoptosis and induced mice subacute liver injury by activating oxidative stress and the ERS-associated PERK pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:177003. [PMID: 39433224 DOI: 10.1016/j.scitotenv.2024.177003] [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: 07/25/2024] [Revised: 09/06/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
With the widespread use of antimony compounds in synthetic materials and processing, the occupational exposure and environmental pollution caused by antimony have attracted the attention of researchers. Studies have shown that antimony compounds can cause liver damage, but the mechanism has not yet been elucidated. In this study, we used the trivalent potassium antimony tartrate (PAT) to infect L02 hepatocytes and Kunming (KM) mice to establish an antimony-induced apoptosis model of L02 cells and a subacute liver injury model of KM mice. We found that PAT exposure caused hepatocyte apoptosis and was accompanied by oxidative stress and endoplasmic reticulum stress (ERS), and the ERS-associated PERK pathway was activated. Further experimental results showed that N-acetyl-l-cysteine (NAC) pretreatment or silencing of the PERK gene in L02 cells reduced PAT-induced apoptosis. The activity of SOD and CAT in treated L02 cells was increased, the malondialdehyde content in L02 cells and liver tissues was decreased, and the content of ERS-related proteins GRP78 and CHOP, as well as the content of PERK-pathway-related proteins p-PERK/PERK, p-eif2α/eif2α and ATF4 protein were significantly reduced. Overall, PAT exposure triggered hepatocyte apoptosis and liver injury by inducing oxidative stress and activating the ERS-associated PERK pathway; however, this effect could be alleviated by NAC intervention or silencing of PERK in hepatocytes.
Collapse
Affiliation(s)
- Hualing Zhang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Meng Tang
- Center for Disease Control and Prevention, Jiulongpo District, Chongqing 400050, PR China
| | - Qin Liu
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Die Wu
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Bing Sun
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Jingbang Dong
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Lan Guan
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Jianlan Luo
- Institute of Geophysical & Geochemical Exploration of Hunan, Changsha 411100, PR China
| | - Ming Zeng
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China.
| |
Collapse
|
10
|
Dike PE, Hwang BJ, Campbell T, Awolowo M, Elliott B, Odero-Marah V. HMGA2 regulates GPX4 expression and ferroptosis in prostate cancer cells. Biochem Biophys Res Commun 2024; 736:150859. [PMID: 39447278 PMCID: PMC11560499 DOI: 10.1016/j.bbrc.2024.150859] [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: 06/05/2024] [Revised: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Prostate cancer (PCa) remains a significant global health burden and an increase in oxidative stress is associated with cancer progression. High Mobility Group A2 (HMGA2), a chromatin architectural protein, increases oxidative stress and promotes sensitivity to ferroptosis inducers, however, the mechanism is unknown. We investigated the role of HMGA2 in GPX4 regulation and the impact on cellular responses to oxidative stress and ferroptosis sensitivity. We conducted UALCAN database analysis, western blot analysis, and lipid peroxidation assays to determine the relationship between HMGA2 and GPX4 and the levels of lipid reactive oxygen species in a panel of PCa cell lines, including an enzalutamide-resistant cancer cell line (C4-2B MDVR). Our results show an inverse relationship between HMGA2 and GPX4 expression with high HMGA2 and low GPX4 expression associated with higher Gleason score and lower survival probability in prostate adenocarcinoma (PRAD) patients, while low/moderate HMGA2 expression is positively associated with increased GPX4 expression and higher survival probability. Cell lines showed a moderately negative but not statistically significant correlation between HMGA2 and GPX4 expression, however, PC3 and DU145 PCa cells display higher lipid peroxides concomitant with higher endogenous levels of HMGA2 and low GPX4. Overexpression of wild-type HMGA2 in LNCaP and 22Rv1 cells leads to higher HMGA2 expression compared to Neo control and is associated with higher SLC7A11 and GPX4 expression, while interestingly truncated HMGA2 overexpression in LNCaP and 22Rv1 cells coincides with higher HMGA2 and reduced GPX4 expression, leading to increased lipid peroxides and susceptibility to ferroptosis. Overexpression of wild-type and truncated HMGA2 in 22Rv1 cells increases SLC7A11 mRNA yet differing GPX4 protein expression suggests posttranslational regulation of GPX4. Moreover, enzalutamide-resistant C4-2B MDVR cells display higher HMGA2 levels compared to C4-2B cells, as well as sensitivity to RSL3 ferroptosis inducer, which is partially reversed by ferroptosis inhibitor, ferrostatin-1. Interestingly, GPX4 expression is higher in C4-2B MDVR cells compared to C4-2B, and HMGA2 knockdown further increases its expression but does not significantly alter its susceptibility to ferroptosis. In conclusion, our study shows that HMGA2 regulation of GPX4 expression is complex and truncated HMGA2 downregulates GPX4 and increases lipid peroxides. Moreover, HMGA2-expressing cells including enzalutamide-resistant cells are susceptible to RSL-3-induced ferroptosis. Thus, ferroptosis sensitivity offers promising insights for the development of targeted therapeutic interventions for aggressive PCa.
Collapse
Affiliation(s)
- Precious Elechi Dike
- Center for Urban Health Disparities Research and Innovation, Department of Biology, Morgan State University, Baltimore, MD, 21251, USA
| | - Bor-Jang Hwang
- Center for Urban Health Disparities Research and Innovation, Department of Biology, Morgan State University, Baltimore, MD, 21251, USA
| | - Taaliah Campbell
- Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, 30314, USA
| | - Mojisoluwa Awolowo
- Center for Urban Health Disparities Research and Innovation, Department of Biology, Morgan State University, Baltimore, MD, 21251, USA
| | - Bethtrice Elliott
- Center for Urban Health Disparities Research and Innovation, Department of Biology, Morgan State University, Baltimore, MD, 21251, USA
| | - Valerie Odero-Marah
- Center for Urban Health Disparities Research and Innovation, Department of Biology, Morgan State University, Baltimore, MD, 21251, USA.
| |
Collapse
|
11
|
Xiao K, Wang S, Li G, Chen W, Chen B, Li X. Resveratrol promotes diabetic wound healing by inhibiting ferroptosis in vascular endothelial cells. Burns 2024; 50:107198. [PMID: 39317551 DOI: 10.1016/j.burns.2024.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/25/2024] [Accepted: 07/04/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Diabetic wounds are a common complication of diabetes, with alarming disability and mortality rates. Ferroptosis plays an essential role in the occurrence and development of diabetes mellitus and its complications, suggesting that mitigating ferroptosis can be used as a potential therapeutic strategy. Resveratrol (RSV) can promote the angiogenesis of diabetic wounds, but its molecular mechanism is unclear, and RSV has a role in regulating ferroptosis. Therefore, we speculated that RSV could promote the angiogenesis of diabetic wounds and accelerate wound healing by regulating ferroptosis. METHODS In this study, we investigated the effects of RSV on human umbilical vein endothelial cells (HUVECs) treated with advanced glycation end-products (AGEs), focusing primarily on cell proliferation and markers associated with ferroptosis. The methods employed included the CCK-8 assay for cell proliferation, ROS determination, Fe²⁺ measurement, scratch and tube formation assays, and transcriptome analysis. To evaluate the effectiveness of RSV in promoting wound healing, we established a type 2 diabetes rat model and created a skin injury model. Wound healing rates were assessed, and tissue samples were analyzed using hematoxylin and eosin (H&E) staining, immunohistochemistry, immunofluorescence, and Western blotting. Additionally, levels of glutathione (GSH) and malondialdehyde (MDA) were measured to evaluate oxidative stress and lipid peroxidation. RESULT Upon treatment of HUVECs with AGEs, we observed a decrease in cell viability and induction of ferroptosis. RSV can alleviate ferroptosis in AGEs-treated HUVECs. Further investigation through transcriptome analysis and Western blotting revealed that RSV alleviates ferroptosis in AGE-treated HUVECs by modulating the activity of nuclear factor erythroid 2-related factor 2 (Nrf2). In vivo experiments using a diabetic rat skin injury model confirmed that both RSV and Ferrostatin-1 (Fer-1) enhance wound healing and angiogenesis. This effect was associated with the regulation of ferroptosis marker proteins including GPX4, SLC7A11, and ACSL4. Additionally, in the diabetic rat groups treated with RSV and Fer-1, we noted increased expression of Nrf2, vascular endothelial growth factor (VEGF), and CD31 proteins compared to the diabetic rat control group. CONCLUSION In diabetic wounds, AGEs can lead to ferroptosis in HUVECs. RSV can inhibit AGE-induced ferroptosis in HUVECs, further promoting angiogenesis in diabetic wounds, and ultimately accelerating wound healing.
Collapse
Affiliation(s)
- Kui Xiao
- Department of Burn and Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510000, Guangdong, China
| | - Sisi Wang
- Department of Burn Surgery, The First People's Hospital of Zhengzhou, Zhengzhou, 450000, Henan, China
| | - Gang Li
- Department of Burn and Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510000, Guangdong, China
| | - Wenxin Chen
- Department of Gynaecology and Obstetrics, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, 421001, Hunan, China
| | - Bin Chen
- Department of Burn and Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510000, Guangdong, China.
| | - Xiaojian Li
- Department of Burn and Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510000, Guangdong, China.
| |
Collapse
|
12
|
Liang W, Ren Y, Wang Y, Chen W, Mo Z, Yang C, Nie K. Xiao-Ban-Xia Decoction Alleviates Chemotherapy-Induced Nausea and Vomiting by Inhibiting Ferroptosis via Activation of The Nrf2/SLC7A11/GPX4 Pathway. Adv Biol (Weinh) 2024; 8:e2400323. [PMID: 39501722 DOI: 10.1002/adbi.202400323] [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: 06/11/2024] [Revised: 10/17/2024] [Indexed: 12/14/2024]
Abstract
Chemotherapy-induced nausea and vomiting (CINV) represents the common gastrointestinal side effect for cancer patients. Xiao-Ban-Xia decoction (XBXD), a classical anti-emetic traditional Chinese medicine formula, is frequently used for the clinical treatment of CINV. This study used a cisplatin-induced rat pica model to explore whether the anti-emetic mechanism of XBXD in treating CINV is related to ferroptosis. The inflammatory damage of the gastrointestinal tract is evaluated by HE staining and ELISA. The degree of ferroptosis are validated by the iron deposition, the levels of ROS, MDA, and GSH, and the ultrastructure of mitochondria in the gastric antrum and ileum. The potential ferroptosis-related targets of XBXD against CINV are screened by network pharmacology and further assessed by Western blot. XBXD significantly decreased the kaolin consumption in rats, and improved the inflammatory pathological damage, with decreased levels of HMGB1, IL-1β, and TNF-α. Furthermore, XBXD significantly suppressed ferroptosis, as indicated by the improvement of iron deposition, mitochondrial abnormalities, and oxidative stress. The network pharmacology and Western blot results indicated that XBXD activated the Nrf2/SLC7A11/GPX4 signaling pathway. This study proved that XBXD activates the Nrf2/SLC7A11/GPX4 signaling pathway, thereby inhibiting ferroptosis, which represents a critical anti-emetic mechanism of XBXD in combatting CINV.
Collapse
Affiliation(s)
- Wan Liang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Yuke Ren
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Yusu Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Weijian Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Ziyao Mo
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Chenglu Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Ke Nie
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| |
Collapse
|
13
|
Yan Y, Yang N, Qin F, Hao Y. Echinacoside Alleviates Metabolic Dysfunction-Associated Steatotic Liver Disease by Inhibiting Ferroptosis via Nrf2/HMOX1 Pathway. Biomedicines 2024; 12:2728. [PMID: 39767635 PMCID: PMC11726887 DOI: 10.3390/biomedicines12122728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/12/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Background: Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic liver disease characterized by hepatic lipid accumulation, and echinacoside (ECH) has demonstrated antioxidant and anti-inflammatory effects across multiple conditions, it has demonstrated hepatoprotective effects. Ferroptosis represents a novel mechanism of cell demise, differing from apoptosis and autophagy. Emerging research indicates that ferroptosis in hepatocytes plays a role in the development of alcoholic liver disease. This study aimed to reveal the effect and potential mechanism of ECH on MASLD. Methods: The effect of ECH on the viability, lipid deposition, lipid peroxidation, mitochondrial of OA/PA-treated HepG2 cells were evaluated by Cell Counting Kit-8 assay, JC-1 and immunofluorescence assay. Meanwhile, the mechanism of ECH was assessed using transmission electron microscopy and immunofluorescence analysis. Moreover, db/db mice, a spontaneous type 2 diabetes mode, were intragastrically administered ECH by 300 mg/kg or an equivalent volume of saline. Body weight, lipids, and liver function were measured. liver pathology was performed. The mechanism of ECH in vivo was analyzed using Western blot and immunofluorescence analysis in db/db mice. Results: ECH attenuated lipid deposition, lipid peroxidation and ferroptosis induced by OA/PA in HepG2 cells. Mitochondrial morphology and function in HepG2 cells were also preserved by ECH. In db/db mice model of MASLD, ECH markedly ameliorated liver hepatocellular ballooning, inflammatory cell infiltration in the portal area, and fibrous tissue proliferation. ECH also increased the expression of Nrf2, HMOX-1, SLC7A11, and GPX4, and decreased the expression of ACSL4 in liver tissues. Mechanically, ECH repressed ferroptosis by activating the Nrf2/HO-1 signaling pathway. Conclusions: Our research revealed that ECH has the capability to modulate ferroptosis via the Nrf2-HMOX1pathway, consequently mitigating the progression of MASLD. This suggests that ECH has a potential role in the treatment of MASLD.
Collapse
Affiliation(s)
| | | | | | - Yarong Hao
- Department of Geriatric, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan 430060, China; (Y.Y.); (N.Y.); (F.Q.)
| |
Collapse
|
14
|
Jin Y, Tan M, Yin Y, Lin C, Zhao Y, Zhang J, Jiang T, Li H, He M. Oroxylin A alleviates myocardial ischemia-reperfusion injury by quelling ferroptosis via activating the DUSP10/MAPK-Nrf2 pathway. Phytother Res 2024; 38:5290-5308. [PMID: 39225191 DOI: 10.1002/ptr.8315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/30/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Reperfusion therapy is the primary treatment strategy for acute myocardial infarction (AMI). Paradoxically, it can lead to myocardial damage, namely myocardial ischemia/reperfusion injury (MIRI). This study explored whether oroxylin A (OA) protects the myocardium after MIRI by inhibiting ferroptosis and the underlying mechanism. In vivo, we established an MIRI model to investigate the protective effect of OA. In vitro, H9C2 cells were used to explore the regulation of ferroptosis by OA through immunofluorescence staining, western blotting, assay kits, etc. Additionally, RNA sequencing analysis (RNA-seq) and network pharmacology analyses were conducted to elucidate the molecular mechanisms. Our results showed that MIRI caused cardiac structural and functional damage in rats. MIRI promoted ferroptosis, which was consistently observed in vitro. However, pretreatment with OA reversed these effects. The mitogen-activated protein kinases (MAPK) signaling pathway participated in the MIRI process, with dual-specificity phosphatase 10 (DUSP10) found to regulate it. Further confirmation was provided by knocking down DUSP10 using small interfering RNA (siRNA), demonstrating the activation of the DUSP10/MAPK-Nrf2 pathway by OA to protect H9C2 cells from ferroptosis. Our research has demonstrated the mitigating effect of OA on MIRI and the improvement of myocardial function for the first time. The inhibition of ferroptosis has been identified as one of the mechanisms through which OA exerts its myocardial protective effects. Moreover, we have first unveiled that DUSP10 serves as an upstream target involved in mediating ferroptosis, and the regulation of the DUSP10/MAPK-Nrf2 pathway by OA is crucial in inhibiting ferroptosis to protect the myocardium.
Collapse
Affiliation(s)
- Yifeng Jin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
- Department of General Practice, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Mingyue Tan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
- Department of Geriatrics, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
| | - Yunfei Yin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Chen Lin
- Jinjihu Business District Squadron, Suzhou Industrial Park Food and Drug Safety Inspection Team, Suzhou, Jiangsu, P. R. China
| | - Yongjian Zhao
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Jun Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| | - Mingqing He
- Department of Gerontology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P. R. China
| |
Collapse
|
15
|
Li W, Zheng L, Luo P, Chen T, Zou J, Chen Q, Cheng L, Gan L, Zhang F, Qian B. Critical role of non-coding RNA-mediated ferroptosis in urologic malignancies. Front Immunol 2024; 15:1486229. [PMID: 39544949 PMCID: PMC11560455 DOI: 10.3389/fimmu.2024.1486229] [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: 08/27/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024] Open
Abstract
Urologic malignancies, characterized by their high aggressiveness and metastatic potential, pose a significant public health challenge globally. Ferroptosis, a novel mode of cell death, typically arises from intracellular iron ion overload and the accumulation of lipid peroxides. This process has been shown to play a crucial regulatory role in various pathological conditions, particularly in cancer, including urologic cancers. However, the comprehensive regulatory mechanisms underlying ferroptosis remain poorly understood, which somewhat limits its broader application in cancer therapy. Non-coding RNAs (ncRNAs), which encompass microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), are non-coding transcripts that play pivotal roles in various physiological processes, such as proliferation, differentiation, apoptosis, and cell cycle regulation, by modulating the expression of target genes. The biological functions and potential regulatory mechanisms of ncRNAs in the context of cancer-related ferroptosis have been partially elucidated. Research indicates that ncRNAs can influence the progression of urologic cancers by affecting cell proliferation, migration, and drug resistance through the regulation of ferroptosis. Consequently, this review aims to clarify the functions and mechanisms of the ncRNA-ferroptosis axis in urologic cancers and to evaluate the clinical significance of ferroptosis-related ncRNAs, thereby providing new insights into cancer biology and therapeutic strategies that may ultimately benefit a diverse range of cancer patients.
Collapse
Affiliation(s)
- Wei Li
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Liying Zheng
- Department of Graduate, The First Affiliated Hospital of Gannan Medical University, Jiangxi, Jiangxi, China
| | - Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Jun Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Qi Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Le Cheng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Lifeng Gan
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Fangtao Zhang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Biao Qian
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| |
Collapse
|
16
|
Lai Z, Wei Y, He M, Lin C, Ouyang W, Liu X. Toxicity and related molecular mechanisms of Sb(III) in the embryos and larvae of zebrafish (Danio rerio). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 359:124551. [PMID: 39004205 DOI: 10.1016/j.envpol.2024.124551] [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: 05/12/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/16/2024]
Abstract
Antimony (Sb) pollution poses a severe threat to humans and ecosystems due to the extensive use of Sb in various fields. However, little is known about the toxic effects of Sb and its aquatic ecotoxicological mechanism. This study aimed to reveal the toxicity and related molecular mechanisms of trivalent Sb (Sb(III)) in zebrafish embryos/larvae. Sb(III) accumulated in larvae, which correlated with the exposure concentration. Although no significant lethal or teratogenic effects were observed, normal growth and development were affected. Exposure to 10 or 20 mg/L Sb(III) increased the levels of reactive oxygen species in the larvae while enhancing catalase activity and increasing cell apoptosis. Transcriptomic analysis revealed that Sb(III) promoted glutathione metabolism and the ferroptosis pathway. In addition, symptoms associated with ferroptosis, including mitochondrial damage, biochemical levels of related molecules and increased tissue iron content, were detected. Quantitative polymerase chain reaction (qPCR) analyses further confirmed that Sb(III) significantly altered the transcription levels of genes related to the ferroptosis pathway by disrupting iron homeostasis. Furthermore, ferrostatin-1 (Fer-1) mitigated the toxic effects induced by Sb(III) in zebrafish. Our research fills the gap in the literature on the toxicity and mechanism of Sb(III) in aquatic organisms, which is highly important for understanding the ecological risks associated with Sb.
Collapse
Affiliation(s)
- Ziyang Lai
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| | - Yihan Wei
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| | - Mengchang He
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China.
| | - Chunye Lin
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| | - Wei Ouyang
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China; Advanced Interdisciplinary Institute of Environment and Ecology, Beijing Normal University, Zhuhai, 519087, China
| | - Xitao Liu
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|
17
|
Wang L, Wang L. Long noncoding RNA DDX11-AS1 represses sorafenib-induced ferroptosis in hepatocellular carcinoma cells via Nrf2-Keap1 pathway. Discov Oncol 2024; 15:544. [PMID: 39390130 PMCID: PMC11467156 DOI: 10.1007/s12672-024-01431-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
Sorafenib, a first-line therapeutic option for advanced hepatocellular carcinoma (HCC), faces a formidable challenge in the form of emerging resistance. Recently, the oncogene DDX11 antisense RNA 1 (DDX11-AS1) has been implicated in various cancers, including HCC. However, its role in sorafenib resistance remains unknown. Our findings reveal that DDX11-AS1 is upregulated in sorafenib-resistant HCC cells, contributing to their resistance by suppressing ferroptosis. Further investigation elucidated the mechanism by which DDX11-AS1 activates the antioxidant Nrf2-Keap1 pathway. By interacting with Nrf2 and hindering its association with Keap1, DDX11-AS1 enhances the stability and nuclear translocation of Nrf2. In summary, our study unveils the potent role of DDX11-AS1 as an enhancer of sorafenib resistance, inhibiting sorafenib-induced ferroptosis through the activation of the Nrf2-Keap1 pathway in HCC. These findings offer a promising therapeutic strategy to overcome resistance and effectively treat HCC.
Collapse
Affiliation(s)
- Liang Wang
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China, NO. 467, Zhongshan Road, Shahekou district, Liaoning, 116000.
| | - Liming Wang
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China, NO. 467, Zhongshan Road, Shahekou district, Liaoning, 116000.
| |
Collapse
|
18
|
Ma W, Jiang X, Jia R, Li Y. Mechanisms of ferroptosis and targeted therapeutic approaches in urological malignancies. Cell Death Discov 2024; 10:432. [PMID: 39384767 PMCID: PMC11464522 DOI: 10.1038/s41420-024-02195-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024] Open
Abstract
The prevalence of urological malignancies remains a significant global health concern, particularly given the challenging prognosis for patients in advanced disease stages. Consequently, there is a pressing need to explore the molecular mechanisms that regulate the development of urological malignancies to discover novel breakthroughs in diagnosis and treatment. Ferroptosis, characterized by iron-ion-dependent lipid peroxidation, is a form of programmed cell death (PCD) distinct from apoptosis, autophagy, and necrosis. Notably, lipid, iron, and glutathione metabolism intricately regulate intracellular ferroptosis, playing essential roles in the progression of various neoplasms and drug resistance. In recent years, ferroptosis has been found to be closely related to urological malignancies. This paper provides an overview of the involvement of ferroptosis in the pathogenesis and progression of urological malignancies, elucidates the molecular mechanisms governing its regulation, and synthesizes recent breakthroughs in diagnosing and treating these malignancies. We aim to provide a new direction for the clinical treatment of urological malignancies.
Collapse
Affiliation(s)
- Wenjie Ma
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xiaotian Jiang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
19
|
Basaran B, Turk H. The levels, single and multiple health risk assessment of 23 metals in enteral nutrition formulas. Food Chem Toxicol 2024; 192:114914. [PMID: 39127122 DOI: 10.1016/j.fct.2024.114914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Enteral nutrition formulas are products that provide macro and micronutrients to patients who cannot receive their nutrition orally. In this study, the levels of 23 metals known to have potential health risks were determined by inductively coupled plasma mass spectrometry in a total of 28 enteral nutrition formula. Metal exposure was calculated according to three different daily energy intake scenarios (Scenario 1 = 50% oral nutrition + 50% enteral nutrition formula, Scenario 2 = 25% oral nutrition + 75% enteral nutrition formula and Scenario 3 = 100% enteral nutrition formula) and evaluated in terms of non-carcinogenic health risks. The mean levels of Fe, Co, Ni, Cu, Zn, Mo, Se, Li, Be, V, As, Sr, Ag, Cd, Sb, Ba, La, Hg and Pb in the samples analyzed were determined 12,000 ± 3300, 64 ± 1.6, 10 ± 13, 1300 ± 400, 8500 ± 2500, 75 ± 30, 61 ± 21, 0.34 ± 0.36, 0.05 ± 0.08, 7.3 ± 2, 1.6 ± 0.6, 457 ± 166, 0.02 ± 0.1, 0.14 ± 0.12, 0.01 ± 0.1, 74 ± 103, 0.63 ± 0.4, 0.05 ± 0.03 and 0.14 ± 0.7 μg/L. These metals were considered safe in terms of non-carcinogenic health risks when analyzed individually. However, when the target hazard quotient values of all metals were evaluated together, hazard index values were higher than the reference value of 1, for both men and women, indicating potential health risks.
Collapse
Affiliation(s)
- Burhan Basaran
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Recep Tayyip Erdogan University, Rize, 53100, Türkiye.
| | - Hulya Turk
- Department of Biology, Science Faculty, Ataturk University, Erzurum, 25240, Türkiye
| |
Collapse
|
20
|
Li MY, Shi YC, Xu WX, Zhao L, Zhang AZ. Exploring Cr(VI)-induced blood-brain barrier injury and neurotoxicity in zebrafish and snakehead fish, and inhibiting toxic effects of astaxanthin. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 355:124280. [PMID: 38815890 DOI: 10.1016/j.envpol.2024.124280] [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: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
Cr(VI) is a common hazardous heavy metal contaminant that seriously endangers human and aquatic animal health. GPX4 was the key enzyme that reduces heavy metal toxicity through inhibiting ferroptosis pathway. Astaxanthin was GPX4 activator that can weaken biological toxicity induced by Cr(VI) exposure. The present study was conducted to evaluate the major role of GPX4 in astaxanthin protects Cr(VI)-induced oxidative damage, blood-brain barrier injury and neurotoxicity in brain-liver axis through inhibiting ferroptosis pathway. In the current study, astaxanthin intervention can effectively alleviate Cr(VI)-induced oxidative stress, blood-brain barrier damage, and neurotoxicity. GPX4 plays a major role in mediating astaxanthin nutritional intervention to reduce ROS and liver non-heme iron accumulation, which would contribute to the reduction of ferroptosis. Meanwhile, astaxanthin maintains the stability of transport receptors and protein macromolecules such as TMEM163, SLC7A11, SLC3A2, FPN1 and GLUT1 in the brain liver axis, promoting substance exchange and energy supply. Moreover, astaxanthin alleviates Cr(VI)-induced neurotoxicity by promoting tight protein expression and reducing blood-brain barrier permeability.
Collapse
Affiliation(s)
- Mu-Yang Li
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Yan-Chao Shi
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Wan-Xin Xu
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Lei Zhao
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China.
| | - Ai-Zhong Zhang
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China.
| |
Collapse
|
21
|
Wu L, Hu Z, Song XF, Liao YJ, Xiahou JH, Li Y, Zhang ZH. Targeting Nrf2 signaling pathways in the role of bladder cancer: From signal network to targeted therapy. Biomed Pharmacother 2024; 176:116829. [PMID: 38820972 DOI: 10.1016/j.biopha.2024.116829] [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: 10/03/2023] [Revised: 05/09/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024] Open
Abstract
Bladder cancer (BC) is the most common malignancy of the urinary system and often recurs after tumor removal and/or is resistant to chemotherapy. In cancer cells, the activity of the signaling pathway changes significantly, affecting a wide range of cell activities from growth and proliferation to apoptosis, invasion and metastasis. Nrf2 is a transcription factor that plays an important role in cellular defense responses to a variety of cellular stresses. There is increasing evidence that Nrf2 acts as a tumor driver and that it is involved in the maintenance of malignant cell phenotypes. Abnormal expression of Nrf2 has been found to be common in a variety of tumors, including bladder cancer. Over-activation of Nrf2 can lead to DNA damage and the development of bladder cancer, and is also associated with various pathological phenomena of bladder cancer, such as metastasis, angiogenesis, and reduced toxicity and efficacy of therapeutic anticancer drugs to provide cell protection for cancer cells. However, the above process can be effectively inhibited or reversed by inhibiting Nrf2. Therefore, Nrf2 signaling may be a potential targeting pathway for bladder cancer. In this review, we will characterize this signaling pathway and summarize the effects of Nrf2 and crosstalk with other signaling pathways on bladder cancer progression. The focus will be on the impact of Nrf2 activation on bladder cancer progression and current therapeutic strategies aimed at blocking the effects of Nrf2. To better determine how to promote new chemotherapy agents, develop new therapeutic agents, and potential therapeutic targets.
Collapse
Affiliation(s)
- Liang Wu
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China.
| | - Zhao Hu
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Xiao-Fen Song
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Yu-Jian Liao
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Jiang-Huan Xiahou
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Yuan Li
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Zhong-Hua Zhang
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China.
| |
Collapse
|
22
|
Hushmandi K, Einollahi B, Saadat SH, Lee EHC, Farani MR, Okina E, Huh YS, Nabavi N, Salimimoghadam S, Kumar AP. Amino acid transporters within the solute carrier superfamily: Underappreciated proteins and novel opportunities for cancer therapy. Mol Metab 2024; 84:101952. [PMID: 38705513 PMCID: PMC11112377 DOI: 10.1016/j.molmet.2024.101952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND Solute carrier (SLC) transporters, a diverse family of membrane proteins, are instrumental in orchestrating the intake and efflux of nutrients including amino acids, vitamins, ions, nutrients, etc, across cell membranes. This dynamic process is critical for sustaining the metabolic demands of cancer cells, promoting their survival, proliferation, and adaptation to the tumor microenvironment (TME). Amino acids are fundamental building blocks of cells and play essential roles in protein synthesis, nutrient sensing, and oncogenic signaling pathways. As key transporters of amino acids, SLCs have emerged as crucial players in maintaining cellular amino acid homeostasis, and their dysregulation is implicated in various cancer types. Thus, understanding the intricate connections between amino acids, SLCs, and cancer is pivotal for unraveling novel therapeutic targets and strategies. SCOPE OF REVIEW In this review, we delve into the significant impact of amino acid carriers of the SLCs family on the growth and progression of cancer and explore the current state of knowledge in this field, shedding light on the molecular mechanisms that underlie these relationships and highlighting potential avenues for future research and clinical interventions. MAJOR CONCLUSIONS Amino acids transportation by SLCs plays a critical role in tumor progression. However, some studies revealed the tumor suppressor function of SLCs. Although several studies evaluated the function of SLC7A11 and SLC1A5, the role of some SLC proteins in cancer is not studied well. To exert their functions, SLCs mediate metabolic rewiring, regulate the maintenance of redox balance, affect main oncogenic pathways, regulate amino acids bioavailability within the TME, and alter the sensitivity of cancer cells to therapeutics. However, different therapeutic methods that prevent the function of SLCs were able to inhibit tumor progression. This comprehensive review provides insights into a rapidly evolving area of cancer biology by focusing on amino acids and their transporters within the SLC superfamily.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
23
|
Gong G, Wan Y, Liu Y, Zhang Z, Zheng Y. Ononin triggers ferroptosis-mediated disruption in the triple negative breast cancer both in vitro and in vivo. Int Immunopharmacol 2024; 132:111959. [PMID: 38554442 DOI: 10.1016/j.intimp.2024.111959] [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: 02/09/2024] [Revised: 03/16/2024] [Accepted: 03/26/2024] [Indexed: 04/01/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is difficult to treat due to a lack of targeted therapies. In this study, we aimed to investigate whether a natural flavonoid compound called ononin could be effective in treating TNBC by triggering ferroptosis in MDA-MB-231 and 4 T1 cell lines, and MDA-MB-231-xenograft nude mice model. Ononin inhibited TNBC through ferroptosis, which was determined by MTT assay, flow cytometry, RT-PCR, immunofluorescence, transmission electron microscopy, histological analysis, western blot and bioluminescence assay. Our results showed that treatment with ononin led to increased levels of malondialdehyde and reactive oxygen species and decreased activity of superoxide dismutase, which are indicatives of ferroptosis. We also found that ononin downregulated two key markers of ferroptosis, SLC7A11 and Nrf2, at both the transcriptional and translational level. Additionally, the administration of ononin resulted in a notable decrease in tumor size and weight in the mouse model. Furthermore, it was observed to enhance the rate of apoptosis in TNBC cells. Importantly, ononin did not induce any histological changes in the kidney, liver, and heart. Taken together, our findings suggest that ononin could be a promising therapeutic strategy for TNBC, and that it works by disrupting the Nrf2/SLC7A11 axis through ferroptosis. These results are encouraging and may lead to the development of new treatments for this challenging cancer subtype.
Collapse
Affiliation(s)
- Guowei Gong
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong 519041, China; Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China.
| | - Yukai Wan
- Second Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Yaqun Liu
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Zhenxia Zhang
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Yuzhong Zheng
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China.
| |
Collapse
|
24
|
Gong G, Ganesan K, Liu Y, Huang Y, Luo Y, Wang X, Zhang Z, Zheng Y. Danggui Buxue Tang improves therapeutic efficacy of doxorubicin in triple negative breast cancer via ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117655. [PMID: 38158099 DOI: 10.1016/j.jep.2023.117655] [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: 11/16/2023] [Revised: 12/14/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danggui Buxue Tang (DBT) has been used for over 800 years to enhance Qi and nourish Blood, and it is particularly beneficial for cancer patients. Recent research has shown that combining DBT with chemotherapy agents leads to superior anti-cancer effects, thereby enhancing therapeutic efficacy. AIM OF THE STUDY The aim of this study was to evaluate the effectiveness of a combination therapy involving doxorubicin (DOX) and Danggui Buxue Tang (DBT) in the treatment of triple-negative breast cancer (TNBC) and to elucidate the underlying mechanisms of action. MATERIALS AND METHODS In vitro experiments were performed using MDA-MB-231 and 4T1 cells, while in vivo experiments were carried out using MDA-MB-231 xenograft mice. The therapeutic effects of the combination therapy were evaluated using various techniques, including MTT assay, colony formation assay, flow cytometry, transwell assay, immunofluorescence, transmission electron microscopy (TEM), histological analysis, western blotting, and bioluminescence assay. RESULTS DBT was found to enhance DOX's anti-TNBC activity in vitro by promoting ferroptosis, as evidenced by the observed mitochondrial morphological changes using TEM. The combination therapy was also found to reduce the expression of Nrf2, HO-1, and GPX4, which are all targets for ferroptosis induction, while simultaneously increasing ROS production. Additionally, the combination therapy reduced nuclear accumulation and constitutive activation of Nrf2, which is a significant cause of chemotherapy resistance and promotes cancer growth. In vivo experiments using an MDA-MB-231 xenograft animal model revealed that the combination therapy significantly reduced tumor cell proliferation and accelerated TNBC deaths by modulating the Nrf2/HO-1/GPX4 axis, with no evidence of tissue abnormalities. Moreover, the combination therapy exhibited a liver protective effect, and administration of Fer-1 was able to reduce the ROS formation produced by the DBT + DOX combination therapy. CONCLUSION This study provides evidence that the combination therapy of DOX and DBT has the potential to treat TNBC by promoting ferroptosis through the Nrf2/HO-1/GPX4 axis.
Collapse
Affiliation(s)
- Guowei Gong
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong, 519041, China; Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China.
| | - Kumar Ganesan
- School of Chinese Medicine, The Hong Kong University, Hong Kong SAR, 999077, China
| | - Yaqun Liu
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Yongping Huang
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Yuting Luo
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Xuexu Wang
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Zhenxia Zhang
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Yuzhong Zheng
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China; Guangdong East Drug and Food & Health Branch, Chaozhou, Guangdong, 521041, China.
| |
Collapse
|
25
|
Long J, Wang W, Chu J, Li Y, Wang M, Su J, Yang Y, Wang G, Li Q, Cheng H. Overexpression of Nrf2 reverses ferroptosis induced by Arenobufagin in gastric cancer. Toxicol Appl Pharmacol 2024; 484:116842. [PMID: 38307257 DOI: 10.1016/j.taap.2024.116842] [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: 11/05/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/04/2024]
Abstract
Arenobufagin (ArBu) is a natural monomer extracted and isolated from the secretion of the Chinese toad, also known as toad venom. This compound exerts anti-tumor effects by promoting apoptosis in tumor cells, inhibiting tumor angiogenesis, and preventing the invasion and migration of tumor cells. However, their impact on ferroptosis in tumor cells has yet to be fully confirmed. In this study, we established a subcutaneous transplant tumor model in nude mice to investigate the inhibitory effect of ArBu on gastric cancer cells (MGC-803) and the safety of drug delivery. in vitro experiments, we screened the most sensitive cancer cell lines using the MTT method and determined the response of ArBu to cell death. Use flow cytometry to measure cytoplasmic and lipid reactive oxygen species (ROS) levels. Determine the expression levels of ferritin-related proteins through Western blot experiments. In addition, a MGC-803 cell model overexpressing Nrf2 was created using lentiviral transfection to investigate the role of ArBu in inducing ferroptosis in cancer cells. Our research findings indicate that ArBu inhibits the proliferation of MGC-803 cells and is linked to ferroptosis. In summary, our research findings indicate that ArBu is a potential anti-gastric cancer drug that can induce ferroptosis in human cancer cells through the Nrf2/SLC7A11/GPX4 pathway.
Collapse
Affiliation(s)
- Jiao Long
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road, Shushan District, Hefei 230038, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wenjun Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road, Shushan District, Hefei 230038, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jing Chu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road, Shushan District, Hefei 230038, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yueyue Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road, Shushan District, Hefei 230038, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Meng Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road, Shushan District, Hefei 230038, China
| | - Jingjing Su
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road, Shushan District, Hefei 230038, China
| | - Yuting Yang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road, Shushan District, Hefei 230038, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - GuoKai Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Qinglin Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road, Shushan District, Hefei 230038, China.
| | - Hui Cheng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road, Shushan District, Hefei 230038, China.
| |
Collapse
|