1
|
Yang T, Yang X, Wang G, Jia D, Li Y. Unraveling the crucial role of SDF-1 in osteoarthritis progression: IL6/HIF-1α positive feedback and chondrocyte ferroptosis. Int Immunopharmacol 2025; 152:114400. [PMID: 40058106 DOI: 10.1016/j.intimp.2025.114400] [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: 01/07/2025] [Revised: 02/15/2025] [Accepted: 02/27/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND Osteoarthritis (OA) is a common joint disease with an incompletely understood pathogenesis. SDF-1, a key factor in cartilage matrix degradation, is involved in OA cartilage degeneration, yet its mechanism, especially regarding ferroptosis, remains unclear. This study focuses on elucidating the role of SDF-1-induced chondrocyte ferroptosis and the IL6/HIF-1α signalling axis in OA. METHODS A rabbit OA model was created via SDF-1 induction. Knee cartilage tissues were sequenced and analyzed bioinformatically to identify key genes, and explore critical pathways. Clinical tissue samples were utilized to validate their clinical relevance. Furthermore, cell and rabbit models were constructed through gene interference and pathway blocking. The expression of related genes and proteins was detected by QPCR, ELISA, Western blot, and immunofluorescence. Additionally, OA and ferroptosis indicators such as cell viability, immunohistochemistry, ROS, lipid ROS, Fe2+, MDA, and mitochondrial morphology were evaluated to uncover the molecular mechanism by which SDF-1 regulates the IL6/HIF-1α signalling axis to mediate chondrocyte ferroptosis. RESULTS Bioinformatics revealed that ferroptosis was significantly activated in SDF-1-induced OA, with IL6 and HIF-1 pathways implicated. In vitro and in vivo, SDF-1 increased the expression and secretion of MMP13 but decreased COL2A1 and ACAN in chondrocytes, leading to OA-like changes. It also suppressed the expression levels of SLC7A11 and GPX4, upregulated the gene and protein levels of ACSL4, promoted the accumulation of MDA, Fe2+, and ROS, and caused mitochondrial morphological changes. These ferroptosis manifestations could be alleviated by the ferroptosis inhibitor Fer-1. IL6 was an important mediator of SDF-1-induced ferroptosis, and knocking down IL6 also inhibited chondrocyte ferroptosis changes. Overexpressing IL6 (oeIL6) and using PX478 to inhibit the HIF-1 signalling pathway showed that PX478 could significantly relieve the cytotoxicity produced by the culture of oeIL6 and SDF-1, enhance chondrocyte viability, reverse the decreased expression of SLC7A11 and GPX4 caused by oeIL6, increase the expression of ACSL4, reverse the accumulation of MDA, Fe2+, and ROS. Moreover, PX478 could also significantly reduce the expression and secretion of IL6. CONCLUSION SDF-1 mediates chondrocyte ferroptosis via the IL6/HIF-1α positive feedback, promoting OA.
Collapse
Affiliation(s)
- Tengyun Yang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Xianguang Yang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Guoliang Wang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Di Jia
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Yanlin Li
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China.
| |
Collapse
|
2
|
Zhao Y, Peng M, Liu H, Zhang X, Fu D. Prepubertal Exposure to Tris(2-chloroethyl) Phosphate Disrupts Blood-Testis Barrier Integrity via Ferritinophagy-Mediated Ferroptosis. TOXICS 2025; 13:285. [PMID: 40278601 DOI: 10.3390/toxics13040285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025]
Abstract
Tris(2-chloroethyl) phosphate (TCEP) is a representative chlorinated organophosphate flame retardant (OPFR) that demonstrates greater persistence than other non-halogenated alkyl or aryl OPFRs. Although TCEP has been shown to accumulate significantly in the environment and contribute to testicular toxicity and spermatogenic dysfunction, the precise underlying factors and mechanisms of action remain unclear. Herein, male ICR mice were gavaged with corn oil, 50 mg/kg body weight (bw) TCEP, or 100 mg/kg bw TCEP from postnatal day (PND) 22 to PND 35. TCEP exposure resulted in the disruption of blood-testis barrier (BTB) integrity and in abnormal testicular development. Considering that Sertoli cells constitute the primary target of toxicants and that TCEP induces oxidative stress in the testis and other organs, we focused on ferroptosis in Sertoli cells. Our findings revealed a significant increase in ferroptosis in the testes and Sertoli cells following TCEP exposure, and we observed functional restoration of Sertoli cell junctions upon treatment with the ferroptosis inhibitor ferrostatin-1. Furthermore, ferritin heavy chain 1 (FTH1) was markedly reduced in TCEP-exposed testes and Sertoli cells. Since nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy is essential for the degradation of FTH1, we assessed ferritinophagic activity and found significant upregulation of NCOA4, ATG5, ATG7, and LC3B II/I in TCEP-exposed testes and Sertoli cells. These results strongly suggest that TCEP triggers Sertoli cell ferroptosis by activating ferritinophagy that leads to reduced expression of BTB-associated proteins, ultimately causing BTB disruption and testicular developmental toxicity.
Collapse
Affiliation(s)
- Yonggang Zhao
- Environment Monitoring Center of Jiangsu Province, Nanjing 210019, China
| | - Mo Peng
- Environment Monitoring Center of Jiangsu Province, Nanjing 210019, China
| | - Honglei Liu
- Nanjing Shenghong Environmental Technology Co., Ltd., Nanjing 210017, China
| | - Xiaoyu Zhang
- Environment Monitoring Center of Jiangsu Province, Nanjing 210019, China
| | - Dan Fu
- Environment Monitoring Center of Jiangsu Province, Nanjing 210019, China
| |
Collapse
|
3
|
Lv H, Mu B, Xu H, Li X, Yao X, Wang Q, Yang H, Ding J, Wang J. Do emerging alternatives pose similar soil ecological risks as traditional plasticizers? A multi-faceted analysis using earthworms as a case study. JOURNAL OF HAZARDOUS MATERIALS 2025; 487:137298. [PMID: 39847925 DOI: 10.1016/j.jhazmat.2025.137298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/05/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
The extensive application of plasticizers has led to significant environmental issues. This study focused on the ecotoxic effects on earthworms of the traditional plasticizer di(2-ethylhexyl) phthalate (DEHP) and non-phthalate plasticizers di(ethylhexyl) terephthalate (DEHT) and acetyltributyl citrate (ATBC). At an environmentally relevant concentration (50 mg/kg), significant accumulation of ROS was observed in earthworms, with a trend of DEHP > DEHT > ATBC, inducing oxidative stress and lipid peroxidation. DEHP, DEHT, and ATBC impaired the energy metabolism in earthworms, as evidenced by a sharp reduction in ATP content ranging from 43.2 % to 75.8 %, which was attributed to the disruption of glycolysis and the TCA cycle. Concurrently, the numbers of cocoons and juvenile earthworms decreased by 23.3 %-76.7 % and 24.2 %-75.8 %, respectively, indicating a significant decline in reproductive capacity. Using qPCR, AlphaFold2, and molecular docking techniques, this study is the first to report that because of their similar molecular structures, the alternatives to DEHP exhibit estrogen-like effects in earthworms, which may be a key mechanism of reproductive toxicity. These results provide valuable references and profound insights for the development of novel plasticizer alternatives and the assessment of their impact on soil ecosystems.
Collapse
Affiliation(s)
- Huijuan Lv
- College of Resources and Environment, Shandong Agricultural University, Tai'an, Shandong 271018, PR China
| | - Baoyan Mu
- College of Resources and Environment, Shandong Agricultural University, Tai'an, Shandong 271018, PR China
| | - Haixia Xu
- Dongying Ecological Environment Monitoring Centre, Dongying Ecological Environment Bureau, Dongying, Shandong 257000, PR China
| | - Xianxu Li
- College of Resources and Environment, Shandong Agricultural University, Tai'an, Shandong 271018, PR China
| | - Xiangfeng Yao
- College of Resources and Environment, Shandong Agricultural University, Tai'an, Shandong 271018, PR China
| | - Qian Wang
- College of Resources and Environment, Shandong Agricultural University, Tai'an, Shandong 271018, PR China
| | - Huiyan Yang
- College of Resources and Environment, Shandong Agricultural University, Tai'an, Shandong 271018, PR China
| | - Jia Ding
- College of Resources and Environment, Shandong Agricultural University, Tai'an, Shandong 271018, PR China
| | - Jun Wang
- College of Resources and Environment, Shandong Agricultural University, Tai'an, Shandong 271018, PR China.
| |
Collapse
|
4
|
Li M, Liu Q, Xie S, Weng D, He J, Yang X, Liu Y, You J, Liao J, Wang P, Lu X, Zhao J. Transformable Tumor Microenvironment-Responsive Oxygen Vacancy-Rich MnO 2@Hydroxyapatite Nanospheres for Highly Efficient Cancer Sonodynamic Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414162. [PMID: 39960349 PMCID: PMC11984894 DOI: 10.1002/advs.202414162] [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: 11/02/2024] [Revised: 12/31/2024] [Indexed: 04/12/2025]
Abstract
Despite the promise of sonodynamic therapy (SDT)-mediated immunotherapy, the anticancer efficacy of current sonosensitizers is greatly limited by the immunosuppressive tumor microenvironment (TME) and their inability to selectively respond to it. Herein, oxygen vacancy-rich MnO2@hydroxyapatite (Ca10(PO4)6(OH)2) core-shell nanospheres (denoted as Ov-MO@CPO) as an advanced TME-responsive sonosensitizer for sonodynamic immunotherapy is demonstrated. The Ov-MO@CPO maintains its structural integrity under neutral conditions but dissolves the pH-sensitive hydroxyapatite shell under acidic TME to release active oxygen vacancy-rich MnO2 core, which reinvigorates H2O2 consumption and hypoxia alleviation due to its catalase-like activity. Furthermore, the introduced oxygen vacancies optimize the electronic structure of Ov-MO@CPO, with active electronic states near the Fermi level and higher d-band center. It results in accelerated electron-hole pair separation and lower catalytic energy barriers to boost ultrasound (US)-initiated ROS production. These multimodal synergistic effects effectively reverse the immunosuppressive tumor microenvironment, inhibiting tumor growth and metastasis in 4T1 tumor-bearing mice. No evident toxic effects are observed in normal mouse tissues. Additionally, when combined with an immune checkpoint inhibitor, Ov-MO@CPO-mediated SDT further improves the effectiveness of immunotherapy. This work affords a new avenue for developing TME-dependent sonosensitizers for SDT-mediated immunotherapy.
Collapse
Affiliation(s)
- Minxing Li
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Qiyu Liu
- The Key Lab of Low‐carbon Chem & Energy Conservation of Guangdong ProvinceSchool of ChemistrySun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Songzuo Xie
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Desheng Weng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinjun He
- The Key Lab of Low‐carbon Chem & Energy Conservation of Guangdong ProvinceSchool of ChemistrySun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Xinyi Yang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Yuanyuan Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinqi You
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinghao Liao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| | - Peng Wang
- Department of Emergency MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Xihong Lu
- The Key Lab of Low‐carbon Chem & Energy Conservation of Guangdong ProvinceSchool of ChemistrySun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Jingjing Zhao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer Medicine, Department of BiotherapySun Yat‐Sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
5
|
Zheng S, Zhao N, Lin X, Jiang L, Qiu C, Jiang J, Shu Z, Qian Y, Liang B, Qiu L. Fine Particulate Matter ( PM2.5) and the Blood-Testis Barrier: An in Vivo and in Vitro Mechanistic Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2025; 133:47006. [PMID: 40043229 PMCID: PMC12010934 DOI: 10.1289/ehp14447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/10/2024] [Accepted: 01/30/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Fine particulate matter [particulate matter (PM) with aerodynamic diameter of ≤ 2.5 μ m (PM 2.5 )] is considered a major component of ambient PM. Exposure to PM 2.5 was shown to be associated with male reproductive system injury. Ferroptosis is regarded as an iron-dependent programmed cell death that is associated with the pathological process. It has been reported that SIRT1 has protective effects on the male reproductive system. However, the underlying mechanisms of exposure to PM 2.5 -induced testicular injury are still unexplored. OBJECTIVES In this study, we investigated the relationship between ferroptosis and male reproductive injury after exposure to PM 2.5 and the role of SIRT1/HIF-1 α signaling pathway in this process. METHODS We established a PM 2.5 exposure model in vivo and in vitro using Sertoli cell Sirt1 conditional knockout C57BL/6 (cKO) mice testes and primary Sertoli cells. Hematoxylin and eosin (H&E) staining were conducted to examine the histology of the mice testes. Sperm parameters and biotin tracer assay were conducted to evaluate the effects of exposure to PM 2.5 on the mice testes. Related markers and genes related to the blood-testis barrier (BTB) and ferroptosis were measured by quantitative real-time polymerase chain reaction (qPCR), western blot, and immunofluorescence assay. siRNA transfection was used to evaluate the potential mechanism. RESULTS Significant pathological damage and lower sperm quality were detected in mice testes exposed to PM 2.5 . We found that exposure to PM 2.5 damaged the BTB and inhibited the expression level of the BTB-related proteins (including Connexin 43, Occludin, Claudin 11, N-Cadherin and ZO-1). According to the enrichment analysis results, ferroptosis and HIF-1 α signaling pathway were significantly enriched in mice testes and primary Sertoli cells exposed to PM 2.5 . Subsequent experiments were conducted to verify the results of the enrichment analysis and revealed differences in the expression levels of HIF-1 α , ferroptosis-related genes (including GPX4, SLC7A11, ACSL4, and HO-1) and ferroptosis-related markers [including malondialdehyde (MDA), glutathione (GSH), and Fe 2 + ], associated with lower expression of SIRT1 after exposure to PM 2.5 . These results suggest that PM 2.5 exposure may be associated with ferroptosis and HIF-1 α signaling pathway in male reproductive dysfunction. CONCLUSIONS Taken together, in vivo and in vitro experiments verified that PM 2.5 exposure in mice may lead to testicular dysfunction through new pathways. https://doi.org/10.1289/EHP14447.
Collapse
Affiliation(s)
- Shaokai Zheng
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
- People’s Hospital of Chenghai Shantou, Shantou, PR China
| | - Nannan Zhao
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
| | - Xiaojun Lin
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
| | - Lianlian Jiang
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
| | - Chong Qiu
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
| | - Jinchen Jiang
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
| | - Zhenhao Shu
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
| | - Yingyun Qian
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
| | - Bo Liang
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
- Department of Ultrasound, Affiliated Hospital 2 of Nantong University, Nantong, PR China
| | - Lianglin Qiu
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, PR China
| |
Collapse
|
6
|
Zhao P, Zhao Y, Ma Y, Liang C, Yuan Q, Gao Y, Liu X, Zhu X, Hao X, Liang G, Fan H, Wang D. Gestational and lactational exposure to DEHP triggers ACSL4/TFR-mediated hippocampal neuronal ferroptosis via YAP activation: Implication for the neurocognitive disorders in male offspring. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138081. [PMID: 40187248 DOI: 10.1016/j.jhazmat.2025.138081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/07/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is one of the most extensively used phthalate and poses a public health concern. Perinatal exposure to DEHP has been shown to cause neurodevelopmental abnormalities and neurobehavioral disorders in offspring. However, the precise molecular mechanism has not yet been fully elucidated. In this study, pregnant C57BL/6 mice were exposed to DEHP from gestation to weaning. By RNA sequencing and animal experiments, ferroptosis has been identified as the key pathologic process contributing to DEHP-induced hippocampal injury in adult male offspring. In vitro results also showed that Ferrostatin-1 (Fer-1) effectively ameliorated Mono-(2-ethylhexyl) phthalate (MEHP) -induced cell survival via the inhibiting ferroptosis in HT22 cells. Consistently, we found that the expression of ACSL4 and TFR was significantly up-regulated in offspring hippocampi and MEHP-exposed HT22 neurons. However, silencing ACSL4 or knockdown TFR relieved MEHP-induced generation of lipid ROS and cellular iron accumulation, thereby blocking ferroptosis. Mechanistically, ACSL4/TFR-mediated ferroptosis seemed to be a Yes-associated protein (YAP) dependent via TEA domain transcription factor 4 in HT22 neurons. Importantly, treatment with Fer-1, rosiglitazone, and Deferoxamine effectively rescued DEHP-evoked cognitive decline in adult male offspring. Our findings certified that gestational and lactational exposure to DEHP provoked ACSL4/TFR-mediated hippocampal neuronal ferroptosis via YAP activation.
Collapse
Affiliation(s)
- Pu Zhao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Yuhang Zhao
- Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yilu Ma
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Chen Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Quan Yuan
- Henan Province Rongkang Hospital, Luoyang, China
| | - Yufei Gao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoli Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoying Zhu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xueqin Hao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Gaofeng Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China.
| | - Dongmei Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
7
|
Shi H, Zhao XH, Peng Q, Zhou XL, Liu SS, Sun CC, Cao QY, Zhu SP, Sun SY. Green tea polyphenols alleviate di (2-ethylhexyl) phthalate-induced testicular injury in mice via lncRNA-miRNA-mRNA axis†. Biol Reprod 2025; 112:485-500. [PMID: 39658192 DOI: 10.1093/biolre/ioae179] [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: 05/07/2024] [Revised: 11/17/2024] [Accepted: 12/07/2024] [Indexed: 12/12/2024] Open
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a commonly used plasticizer known for its toxic effects on the male reproductive system. Green tea polyphenols (GTPs), recognized for their antioxidant and anti-inflammatory properties, have demonstrated protective effects on various organs, but the mechanisms by which GTPs mitigate DEHP-induced testicular damage remain unclear. Healthy male C57BL/6J mice were divided into five groups: control, DEHP, DEHP + GTP treatment, GTP, and oil groups. Testicular histopathological changes were assessed using hematoxylin-eosin (H&E), periodic acid-Schiff (PAS), and Masson staining. Ultrastructural alterations were examined through transmission electron microscopy. High-throughput sequencing was performed to analyze the expression of mRNA, miRNA, and lncRNA and construct an lncRNA-miRNA-mRNA regulatory network for identifying key regulatory axes. Mice in the DEHP group exhibited significant testicular damage, including reduced sperm count, mitochondrial deformation, and endoplasmic reticulum dilation. GTP treatment notably improved testicular structural integrity, restored sperm count, and alleviated mitochondrial and endoplasmic reticulum damage. Additionally, DEHP significantly increased activated CD8+ T cells, which were reduced with GTP treatment. High-throughput sequencing revealed that GTP treatment exerted protective effects through the regulation of six key lncRNA-miRNA-mRNA axes. GTPs significantly protect against DEHP-induced testicular damage, and the lncRNA-miRNA-mRNA regulatory axes play a potential role in this process.
Collapse
Affiliation(s)
- Heng Shi
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Ave, Guangzhou 510630, China
- Department of Gastroenterology, The Central Hospital of Shaoyang, No. 360, Baoqing Middle Road, Hongqi Street, Daxiang District, Shaoyang 42200, China
| | - Xin-Hai Zhao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Ave, Guangzhou 510630, China
| | - Qin Peng
- Department of Gastroenterology, The Central Hospital of Shaoyang, No. 360, Baoqing Middle Road, Hongqi Street, Daxiang District, Shaoyang 42200, China
| | - Xian-Ling Zhou
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Ave, Guangzhou 510630, China
| | - Si-Si Liu
- Department of Pathology, The Central Hospital of Shaoyang, No. 360, Baoqing Middle Road, Hongqi Street, Daxiang District, Shaoyang 42200, China
| | - Chuan-Chuan Sun
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Ave, Guangzhou 510630, China
| | - Qiu-Yu Cao
- Department of Gynecologic, Jiangmen Hospital Affiliated to Jinan University, No. 30, Huayuan East Road, Jiangmen 529000, China
| | - Shi-Ping Zhu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Ave, Guangzhou 510630, China
| | - Sheng-Yun Sun
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Ave, Guangzhou 510630, China
| |
Collapse
|
8
|
Hui Q, Du X, Li M, Liu S, Wang Z, Song S, Gao Y, Yang Y, Zhou C, Li Y. Mechanisms and targeted prevention of hepatic osteodystrophy caused by a low concentration of di-(2-ethylhexyl)-phthalate. Front Immunol 2025; 16:1552150. [PMID: 40129988 PMCID: PMC11931061 DOI: 10.3389/fimmu.2025.1552150] [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: 12/27/2024] [Accepted: 02/14/2025] [Indexed: 03/26/2025] Open
Abstract
Objectives Hepatic osteodystrophy (HOD) is an important public health issue that severely affects human health. The pathogenesis of HOD is complex, and exposure to environmental pollutants plays an important role. Di-(2-ethylhexyl) phthalate (DEHP) is a persistent environmental endocrine toxicant that is present in many products, and the liver is an important target organ for its toxic effects. Our research aimed to investigate the effects of DEHP on HOD, and to reveal the underlying mechanisms and the potential key preventive approaches. Methods The daily intake EDI of DEHP and bone density indicators for men and women from 2009 to 2018 were screened and organized from the NHANES database to reveal the population correlation between EDI and BMD; C57BL/6 female and male mice were selected to construct an animal model of DEHP induced HOD, exploring the fuchtions and mechanisms of DEHP on osteoporosis; the novel small molecule inhibitor imICA was used to inhibit the process of DEHP induced osteoporosis, further exploring the targeted inhibition pathway of DEHP induced HOD. Results Male and female populations were exposed to a relatively lower concentration of DEHP, and that only the male population exhibited a negative correlation between DEHP exposure and bone mineral density. An in vivo study confirmed that a low dose of DEHP caused liver lesions, disrupted liver function, and induced osteoporosis in male but not female C57BL/6J mice. Regarding the molecular mechanisms, a low dose of DEHP activated the hepatic 14-3-3η/nuclear factor κB (NF-κB) positive feedback loop, which in turn modified the secretory proteome associated with bone differentiation, leading to HOD. Finally, we revealed that targeting the 14-3-3η/ NF-κB feedback loop using our novel 14-3-3η inhibitor (imICA) could prevent DEHP-induced HOD. Conclusion A low dose of DEHP activated the hepatic 14-3-3η/ NF-κB positive feedback loop, which in turn modified the secretory proteome associated with bone differentiation and elevated IL-6 and CXCL1 levels, leading to HOD. Targeted 14-3-3η/NF-κB feedback loop using our novel 14-3-3η inhibitor, imICA, prevented DEHP-induced HOD.
Collapse
Affiliation(s)
- Qinming Hui
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinru Du
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Maoxuan Li
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Sha Liu
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhendong Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Sisi Song
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yancheng Gao
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ye Yang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chunxiao Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yuan Li
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Nie T, Nepovimova E, Wu Q. Circadian rhythm, hypoxia, and cellular senescence: From molecular mechanisms to targeted strategies. Eur J Pharmacol 2025; 990:177290. [PMID: 39863143 DOI: 10.1016/j.ejphar.2025.177290] [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: 10/25/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Cellular senescence precipitates a decline in physiological activities and metabolic functions, often accompanied by heightened inflammatory responses, diminished immune function, and impaired tissue and organ performance. Despite extensive research, the mechanisms underpinning cellular senescence remain incompletely elucidated. Emerging evidence implicates circadian rhythm and hypoxia as pivotal factors in cellular senescence. Circadian proteins are central to the molecular mechanism governing circadian rhythm, which regulates homeostasis throughout the body. These proteins mediate responses to hypoxic stress and influence the progression of cellular senescence, with protein Brain and muscle arnt-like 1 (BMAL1 or Arntl) playing a prominent role. Hypoxia-inducible factor-1α (HIF-1α), a key regulator of oxygen homeostasis within the cellular microenvironment, orchestrates the transcription of genes involved in various physiological processes. HIF-1α not only impacts normal circadian rhythm functions but also can induce or inhibit cellular senescence. Notably, HIF-1α may aberrantly interact with BMAL1, forming the HIF-1α-BMAL1 heterodimer, which can instigate multiple physiological dysfunctions. This heterodimer is hypothesized to modulate cellular senescence by affecting the molecular mechanism of circadian rhythm and hypoxia signaling pathways. In this review, we elucidate the intricate relationships among circadian rhythm, hypoxia, and cellular senescence. We synthesize diverse evidence to discuss their underlying mechanisms and identify novel therapeutic targets to address cellular senescence. Additionally, we discuss current challenges and suggest potential directions for future research. This work aims to deepen our understanding of the interplay between circadian rhythm, hypoxia, and cellular senescence, ultimately facilitating the development of therapeutic strategies for aging and related diseases.
Collapse
Affiliation(s)
- Tong Nie
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
10
|
Elseweidy MM, Harb NG, Ali AA, El-Aziz RMA, Elrashidy RA. Sulforaphane substantially impedes testicular ferroptosis in adult rats exposed to di-2-ethylhexyl phthalate through activation of NRF-2/SLC7A11/GPX-4 trajectory. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3163-3175. [PMID: 39352535 PMCID: PMC11920001 DOI: 10.1007/s00210-024-03440-w] [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: 07/23/2024] [Accepted: 09/07/2024] [Indexed: 03/19/2025]
Abstract
Di-2-ethylhexyl phthalate (DEHP) is a common plasticizer with a deleterious impact on testicular functionality and male fertility. Growing evidence implicates ferroptosis as one of the plausible mechanisms for DEHP-induced testicular injury. Sulforaphane (SFN) is a natural isothiocyanate displaying beneficial effects on testicular injury in several animal models. Herein, we explored the potential protective effect of SFN on testicular ferroptosis and toxicity evoked by DEHP. Adult male Wistar rats were equally distributed into three groups (n = 6/group): (i) CON group; (ii) DEHP group, received DEHP (2 g/kg PO) for 4 weeks; and (iii) DEHP + SFN group, received SFN (10 mg/kg, PO) 1 week prior to DEHP then concurrently with DEHP for further 4 weeks. Compared to CON group, exposure to DEHP caused testicular atrophy, deteriorated testicular architecture, testicular fibrosis, reduced sperm count and motility, higher sperm deformity, and declined serum testosterone level. All these abnormalities were ameliorated by SFN preconditioning. Additionally, pretreatment with SFN reversed the increased aromatase level and upregulated the steroidogenic markers in testes of DEHP-exposed rats. SFN pretreatment also counteracted DEHP-induced oxidative stress and boosted the total antioxidant capacity in testicular tissue via activation of the nuclear factor erythroid 2-related factor 2 (NRF-2) and its downstream target, hemeoxygenase-1 (HO-1). Moreover, SFN preconditioning mitigated DEHP-induced ferroptosis through up-surging SLC7A11, GPX-4, and GSH, while suppressing iron overload and ACSL4-induced lipid peroxidation in testicular tissue of rats. These findings may nominate SFN as a promising protective intervention to alleviate testicular ferroptosis associated with DEHP exposure through activation of NRF-2/SLC7A11/GPX-4 trajectory.
Collapse
Affiliation(s)
- Mohammed M Elseweidy
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Nouran G Harb
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Abdelmoniem A Ali
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Reda M Abd El-Aziz
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Rania A Elrashidy
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
11
|
Wang X, Zhang T, Wang S, Shi H, Dong H, Huang Y, Lai W, Hu Y, Yue C. Bio-nanocomplexes impair iron homeostasis to induce non-canonical ferroptosis in cancer cells. J Nanobiotechnology 2025; 23:121. [PMID: 39972473 PMCID: PMC11837358 DOI: 10.1186/s12951-025-03117-3] [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: 07/18/2024] [Accepted: 01/13/2025] [Indexed: 02/21/2025] Open
Abstract
The targeted elevation of the labile iron pool (LIP) represents the most direct and effective strategy to induce ferroptosis in cancer cells. However, the efficiency of increasing LIP to induce ferroptosis via iron supplementation is controversial due to the iron homeostasis between LIP and storage iron pool, leading to poor effects and serious safety concerns. In this study, a bio-nanocomplex named AbDA-Lim, composed of albumin, polydopamine, and limonene, is prepared to promote LIP and induce non-canonical ferroptosis in cancer cells by destroying the iron balance. Albumin avidity drives AbDA-Lim entering cancer cells. Subsequently, the released polydopamine enhances the expression of HMOX1 to degrade haem and facilitate the transformation of Fe (III) to Fe (II). Meanwhile, limonene reduces glutathione (GSH) levels via inhibiting CBS, thereby, triggering the release of Fe (II) into LIP from its GSH-bound storage state. The augmentation of LIP ultimately triggers non-canonical ferroptosis in cancer cells. Furthermore, the photothermal property of polydopamine augments the synergistic anti-tumor efficiency of AbDA-Lim by incorporating photothermal therapy. This study presents a distinctive, cascading, and biotic strategy for promoting LIP non-canonically to induce ferroptosis.
Collapse
Affiliation(s)
- Xin Wang
- Department of General Surgery, Center of Nutrition and Metabolism of Cancer, Beijing Shijitan Hospital, Key Laboratory of Cancer FSMP for State Market Regulation, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China
| | - Tianyi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China
| | - Shuai Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China
| | - Hanping Shi
- Department of General Surgery, Center of Nutrition and Metabolism of Cancer, Beijing Shijitan Hospital, Key Laboratory of Cancer FSMP for State Market Regulation, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China
| | - Hong Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China
| | - Yanning Huang
- Office of International Cooperation and Exchanges, Central South University, Changsha, 410008, China
| | - Wenjia Lai
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China.
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China.
| | - Chunyan Yue
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
12
|
Yang Y, Liu M, Dong X, Bai J, Shi W, Zhu Q, Liu J, Wang Z, Yi L, Yin X, Ni J, Qu C. Naringin Suppresses CoCl 2-Induced Ferroptosis in ARPE-19 Cells. Antioxidants (Basel) 2025; 14:236. [PMID: 40002420 PMCID: PMC11852185 DOI: 10.3390/antiox14020236] [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: 01/08/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Hypoxic damage to retinal pigment epithelial (RPE) cells and subsequent neovascularization are key factors in the pathogenesis of branch retinal vein occlusion (BRVO). Naringin (NG), a naturally occurring flavanone glycoside, has demonstrated significant antioxidant and anti-neovascular activities. However, the regulatory effects and mechanisms of NG on ferroptosis in BRVO are yet to be explored. Our study aimed to investigate the protective effects of NG on RPE cells under hypoxic stress and to elucidate the underlying molecular mechanisms. Our findings revealed that NG significantly reduced cytotoxicity induced by cobaltous chloride (CoCl2) and also inhibited vascular proliferation in the retina, thereby attenuating choroidal neovascularization. NG pretreatment largely countered the overproduction of reactive oxygen species (ROS) and malondialdehyde (MDA) triggered by hypoxic damage, while also restoring levels of the antioxidants glutathione (GSH) and superoxide dismutase (SOD). Furthermore, NG pretreatment significantly activated the expression of hypoxia-inducible factor-1 alpha (HIF-1α) and its downstream heme oxygenase-1 (HO-1) and NADPH dehydrogenase (NQO1). In conclusion, NG not only inhibits neovascularization but also alleviates inflammation in RPE cells by modulating the HO-1/GPX4 pathway to inhibit ferroptosis. These findings highlight the potential of NG as a promising therapeutic agent for the treatment of BRVO.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jian Ni
- College of Chinese Herbal Medicine, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.Y.); (M.L.); (X.D.); (J.B.); (Q.Z.); (J.L.); (Z.W.); (L.Y.); (X.Y.)
| | - Changhai Qu
- College of Chinese Herbal Medicine, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.Y.); (M.L.); (X.D.); (J.B.); (Q.Z.); (J.L.); (Z.W.); (L.Y.); (X.Y.)
| |
Collapse
|
13
|
Luo M, Jin T, Fang Y, Chen F, Zhu L, Bai J, Ding J. Signaling Pathways Involved in Acute Pancreatitis. J Inflamm Res 2025; 18:2287-2303. [PMID: 40230438 PMCID: PMC11995411 DOI: 10.2147/jir.s485804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/25/2025] [Indexed: 04/16/2025] Open
Abstract
Acute pancreatitis (AP) is a common digestive emergency with high morbidity and mortality. Over the past decade, significant progress has been made in understanding the mechanisms of AP, including oxidative stress, disruptions in calcium homeostasis, endoplasmic reticulum stress, inflammatory responses, and various forms of cell death. This review provides an overview of the typical signaling pathways involved and proposes the latest clinical translation prospects. These strategies are important for the early management of AP, preventing multi-organ injury, and improving the overall prognosis of the disease.
Collapse
Affiliation(s)
- Mengchen Luo
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Ting Jin
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Yi Fang
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Feng Chen
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Lujian Zhu
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| |
Collapse
|
14
|
Liu X, Wang C, Ma Y, Fu L, Luo W, Xu C, Tian Y, Ma M, Mao Y. Transcriptome analysis reveals the molecular mechanisms of neonicotinoid acetamiprid in Leydig cells. Toxicol Ind Health 2025; 41:61-72. [PMID: 39529242 DOI: 10.1177/07482337241300215] [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] [Indexed: 11/16/2024]
Abstract
At present, the reproductive toxicology of neonicotinoids has received greater attention, however, its potential mechanisms are still not fully understood. Acetamiprid (ACE) is a new-generation neonicotinoid and has become a ubiquitous contaminant in the environment. This study aimed to investigate the toxic effects of ACE in TM3 Leydig cells based on transcriptome analysis. The viability and apoptosis of TM3 cells exposed to different concentrations of ACE were assessed by CCK8 and flow cytometry, respectively. After ACE exposure, transcriptome analysis was performed to screen differential expression genes (DEGs), followed by qPCR verification. Results showed that ACE exposure resulted in a time- and dose-dependent decrease in the viability of TM3 cells (p < .05). ACE also exerted a dose-dependent pro-apoptotic effect on TM3 cells. Results of RNA-seq showed that 1477 DEGs were obtained, of which 539 DEGs were up-regulated and 938 DEGs were down-regulated. GO and KEGG analyses of DEGs showed that DNA replication and cell cycle might be the key mechanisms for the cytotoxicity of ACE. qPCR results demonstrated that Mdm2, Cdkn1a (p21) and Gadd45 were significantly increased, and Pcna, Ccna2 (CycA), Ccnb1 (CycB), Ccne1 (CycE), and Cdk1 were significantly decreased, indicating that ACE exposure might promote G1/S and G2/M cell cycle arrest. Additionally, FoxO, p53, and HIF-1 signaling pathways and ferroptosis might play important roles in ACE-induced reproductive toxicity. Collectively, this study provides new perspectives into the mechanism of ACE-induced reproductive toxicity and lays a theoretical foundation for the in-depth study of non-target toxicity mechanisms of neonicotinoid insecticides.
Collapse
Affiliation(s)
- Xun Liu
- Department of Health Inspection and Quarantine, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Ce Wang
- Department of Health Inspection and Quarantine, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Yue Ma
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Linxiang Fu
- Department of Health Inspection and Quarantine, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Wanji Luo
- Department of Health Inspection and Quarantine, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Changjie Xu
- Department of Health Inspection and Quarantine, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Ying Tian
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingyue Ma
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Yaping Mao
- Department of Health Inspection and Quarantine, School of Public Health, Shenyang Medical College, Shenyang, China
| |
Collapse
|
15
|
Wang C, Zhao Y, Peng Y, Chen W, Zhu J, Gu C, Huo R, Ding L, Liu Y, Liu T, Zhang C, Yang W, Wang H, Guo W, Wang B. Juglone induces ferroptotic effect on hepatocellular carcinoma and pan-cancer via the FOSL1-HMOX1 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156417. [PMID: 39923427 DOI: 10.1016/j.phymed.2025.156417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Drug therapy plays an essential role in the management of hepatocellular carcinoma (HCC). Recently, the use of natural products to suppress tumor cells has emerged as a promising direction for drug development. Juglone, a natural compound, exhibits anticancer activities across various cancer types. However, the precise mechanism underlying the anticancer effect of juglone, especially in HCC, remains elusive. PURPOSE This study aimed to investigate the potential inhibitory effects of juglone on HCC and pan-cancer, as well as elucidate the underlying mechanism. METHODS Cell Counting Kit-8 and colony formation assays were used to examine cell proliferation. Transwell and wound healing assays were used to evaluate cell migration. Cell cycle distribution was assessed by flow cytometry. The in vivo effect of juglone on HCC was evaluated by establishing the HCC xenograft mice model. RNA sequencing and inhibitors targeting diverse modes of programmed cell death were applied to uncover the form of juglone-induced cell death. Integrated transcriptomic, and proteomic analyses unveiled the underlying mechanism. The dual-luciferase reporter assay was employed to verify the findings. The pan-cancer value of juglone was assessed using TCGA database analysis and cellular assays. RESULTS Juglone suppressed HCC growth via ferroptosis in vitro and in vivo, which is evidenced by increased levels of iron, lipid peroxidation (LPO), reactive oxygen species (ROS), malondialdehyde (MDA), and decreased levels of glutathione (GSH). Omic analyses, gene silencing and functional analyses showed the upregulated HMOX1 and FOSL1 were the key effector molecule and transcriptional factor in juglone-induced ferroptosis, respectively. The binding site of FOSL1 at the promoter of HMOX1 was identified. Juglone could induce ferroptosis in pan-cancer by activating the FOSL1-HMOX1 axis. CONCLUSION Our findings, for the first time, demonstrate that juglone effectively inhibits tumor growth by inducing FOSL1-HMOX1-dependent ferroptosis, thereby offering a promising strategy for the development of anticancer drugs.
Collapse
Affiliation(s)
- Chuyu Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ying Zhao
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yingfei Peng
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Wei Chen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Jie Zhu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Chenzheng Gu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ran Huo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Lin Ding
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yu Liu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Chunyan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China; Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, PR China; Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, PR China; Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, PR China
| | - Wenjing Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Hao Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China; Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, PR China; Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, PR China; Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, PR China.
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China; Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, PR China; Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, PR China; Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, PR China.
| |
Collapse
|
16
|
Jia Y, Li R, Li Y, Kachler K, Meng X, Gießl A, Qin Y, Zhang F, Liu N, Andreev D, Schett G, Bozec A. Melanoma bone metastasis-induced osteocyte ferroptosis via the HIF1α-HMOX1 axis. Bone Res 2025; 13:9. [PMID: 39814705 PMCID: PMC11735842 DOI: 10.1038/s41413-024-00384-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/11/2024] [Accepted: 10/15/2024] [Indexed: 01/30/2025] Open
Abstract
Osteocytes are the main cells in mineralized bone tissue. Elevated osteocyte apoptosis has been observed in lytic bone lesions of patients with multiple myeloma. However, their precise contribution to bone metastasis remains unclear. Here, we investigated the pathogenic mechanisms driving melanoma-induced osteocyte death. Both in vivo models and in vitro assays were combined with untargeted RNA sequencing approaches to explore the pathways governing melanoma-induced osteocyte death. We could show that ferroptosis is the primary mechanism behind osteocyte death in the context of melanoma bone metastasis. HMOX1 was identified as a crucial regulatory factor in this process, directly involved in inducing ferroptosis and affecting osteocyte viability. We uncover a non-canonical pathway that involves excessive autophagy-mediated ferritin degradation, highlighting the complex relationship between autophagy and ferroptosis in melanoma-induced osteocyte death. In addition, HIF1α pathway was shown as an upstream regulator, providing a potential target for modulating HMOX1 expression and influencing autophagy-dependent ferroptosis. In conclusion, our study provides insight into the pathogenic mechanisms of osteocyte death induced by melanoma bone metastasis, with a specific focus on ferroptosis and its regulation. This would enhance our comprehension of melanoma-induced osteocyte death.
Collapse
Affiliation(s)
- Yewei Jia
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Rui Li
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixuan Li
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katerina Kachler
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xianyi Meng
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andreas Gießl
- Department of Opthalmology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yi Qin
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Fulin Zhang
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ning Liu
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Darja Andreev
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Technische Universität Dresden (TUD), Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
17
|
Alam MS, Maowa Z, Hasan MN. Phthalates toxicity in vivo to rats, mice, birds, and fish: A thematic scoping review. Heliyon 2025; 11:e41277. [PMID: 39811286 PMCID: PMC11731458 DOI: 10.1016/j.heliyon.2024.e41277] [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: 09/25/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
Background Phthalates, a large group of endocrine disruptors, are ubiquitous in the environment and detrimental to human health. This scoping review aimed to summarize the effects of phthalates on laboratory animals relevant to humans, assess toxicity, and analyze mechanisms of toxicity for public health concerns. Methods Articles were retrieved from Google Scholar, PubMed, ScienceDirect, and Web of Science search engines. The search used the term "toxicity of phthalates in vivo, animals or birds or fish." Original research articles published between 2010 and 2024 describing in vivo toxicity in rat, mouse, bird, and fish models, were included. Conversely, articles that did not meet the above criteria were excluded from this scoping review. Two authors independently extracted data using data extraction tools based on themes, while a third arbitrated if consensus was not met. A senior researcher developed the themes, which were further refined through discussions. Data analysis involved quantitative (percentage of studies) and qualitative (content analysis) methods. Results Of the 8180 articles screened, 153 met the inclusion criteria. Most of them were published after 2015 (74.50 %). The scoping review showed that DEHP (56.20 %) and DBP (21.57 %) were the most studied phthalates followed by BBP, DiBP, DMP, DEP, BBOP, and DiNP. Scarce data were available on DnOP, DPHP, DPeP, DUDP, DTDP, DMiP, and DiOP. Interestingly, studies of combinations of two or more phthalates were also present. The main laboratory animals employed were rats (48.37 %) and mice (39.87 %), while the least studied were birds (5.22 %) and fish (6.53 %). Most studies related to testicular toxicity (37.60 %), hepatotoxicity (23.53 %), and ovarian toxicity (18.30 %) investigations, while the rest consisted of neurotoxicity (6.88 %), renal toxicity (6.53 %), and thyroid toxicity studies (4.57 %). Studies focused on oxidative stress (34.64 %), apoptosis (22.22 %), steroid hormone deprivation (20.26 %), lipid metabolism disorder (11.76 %), and immunotoxicity (5.88 %) as mechanisms of toxicity. The most commonly used techniques were H&E, RT-qPCR, ROS assay, WB, IHC, ELISA, RIA, TUNEL, TEM, IFM, FCM, and RNA-seq. Conclusions DEHP and DBP are the most toxic and studied phthalates, while BBP, DiNP, DiBP, DiDP, BBOP, DMP, and DiOP and their combinations require more accurate studies to confirm their toxic effects on human health and mechanisms of action. These will assist policymakers in adopting strategies to minimize public exposure and adverse effects.
Collapse
Affiliation(s)
- Mohammad Shah Alam
- Department of Anatomy and Histology, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, 1706, Bangladesh
| | - Zannatul Maowa
- Department of Anatomy and Histology, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, 1706, Bangladesh
| | - Mohammad Nazmol Hasan
- Department of Statistics, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| |
Collapse
|
18
|
Li XL, Li QM, Zheng YZ, Hu D, Cai XY, Yin K, Qi YY, Cheng ZY, Ning X, Cai Y, Wu W, Lin TY, Xu T, Zhao LL. Di (2-ethylhexyl) phthalate reduces sperm motility by decreasing sperm tail energy supply. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117811. [PMID: 39879685 DOI: 10.1016/j.ecoenv.2025.117811] [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: 08/31/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Di (2-ethylhexyl) phthalate (DEHP) is widespread in the environment. It can impair sperm function through damaging the sperm development process. However, few studies have focused on the sperm tail that is directly related to sperm motility. In this study, we demonstrate that DEHP impedes the conversion of riboflavin in the mitochondrial sheath in the midsection of the sperm tail, resulting in reduced ATP (adenosine triphosphate) synthesis, thereby inhibiting sperm motility. The results of HPLC-MS/MS showed that DEHP metabolites were transported to the testes and epididium, indicating that MEHP (Mono-2-ethylexyl phthalate) could directly affect the espermatozoa of mature. Sperm motility analysis determined that sperm motility decreased with increasing DEHP concentration. The movement of sperm is mainly dependent on the tail motility, which is largely determined by tail structure and energy supply. Electron microscopy images illustrate that there are no observable changes in the basic kinematic structure of the sperm tail. However, DEHP causes a decrease in complex II activity of mitochondrial respiratory chain by interfering with the synthesis of the cofactor FAD (Flavin adenine dinucleotide), which leads to a decrease in ATP concentration. Therefore, DEHP exposure can reduce sperm motility by decreasing sperm tail energy supply. This study exemplifies the importance of the sperm tail in sperm dysfunction caused by environmental pollutants.
Collapse
Affiliation(s)
- Xiao-Lu Li
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Qi-Meng Li
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Yuan-Zhuo Zheng
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Die Hu
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Xiao-Yue Cai
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Ke Yin
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Yin-Yin Qi
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Zi Yu Cheng
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China
| | - Xia Ning
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Yang Cai
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Wei Wu
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Ting-Yuan Lin
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Tao Xu
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China.
| | - Ling-Li Zhao
- Department of Toxicology, School of Public Health; Suzhou Hospital of Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China.
| |
Collapse
|
19
|
Wu SF, Ga Y, Ma DY, Hou SL, Hui QY, Hao ZH. The role of ferroptosis in environmental pollution-induced male reproductive system toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125118. [PMID: 39414070 DOI: 10.1016/j.envpol.2024.125118] [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: 06/30/2024] [Revised: 09/28/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
This article provides a comprehensive review of the toxic effects of environmental pollution on the male reproductive system, with a particular emphasis on ferroptosis, a form of programmed cell death. Research has shown that environmental pollutants, such as heavy metals, pesticide residues, and plastic additives, can disrupt oxidative stress, increasing the production of reactive oxygen species (ROS) in germ cells. This disruption damages cellular lipids, proteins, and DNA, culminating in cell dysfunction or death. Ferroptosis, a cell death pathway closely linked to oxidative stress, is characterized by the accumulation of intracellular iron ions and elevated levels of lipid ROS. This review also explores the role of ferroptosis in male reproductive disorders, including its contributions to reduced sperm count, decreased motility, and abnormal morphology. Environmental pollutants, particularly heavy metals, can induce ferroptosis by interfering with intracellular antioxidant systems, notably the NRF2, GSH, and GPX4 pathways, accumulating toxic lipid peroxides. Furthermore, the article examines the potential interplay between ferroptosis and other forms of cell death, such as apoptosis, autophagy, pyroptosis, and necrosis, in the context of male reproductive health. The review underscores the critical need for further research into the link between environmental pollutants and male fertility, particularly focusing on ferroptosis. It advocates for targeted research efforts to mitigate the adverse effects of ferroptosis and protect reproductive health, emphasizing that a deeper understanding of these mechanisms could lead to innovative preventive strategies against environmental threats to fertility.
Collapse
Affiliation(s)
- Shao-Feng Wu
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Yu Ga
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Dan-Yang Ma
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Si-Lu Hou
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Qiao-Yue Hui
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Zhi-Hui Hao
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China; Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China; National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China.
| |
Collapse
|
20
|
Xiao S, Cui J, Cao Y, Zhang Y, Yang J, Zheng L, Zhao F, Liu X, Zhou Z, Liu D, Wang P. Adolescent exposure to organophosphate insecticide malathion induces spermatogenesis dysfunction in mice by activating the HIF-1/MAPK/PI3K pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125209. [PMID: 39476999 DOI: 10.1016/j.envpol.2024.125209] [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: 08/19/2024] [Revised: 10/19/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024]
Abstract
Chemical-caused reproductive dysfunction has emerged as a global public health concern. This study investigated the adverse effects of the organophosphorus pesticide malathion on reproductive function in adolescent male mice at environmentally relevant concentrations. The results indicated that eight-week malathion exposure reduced testis weight, caused sex and thyroid hormone disorders, and induced testicular spermatogenic epithelium damage and oxidative stress. Testicular RNA sequencing indicated that malathion significantly affected testicular energy metabolism, hypoxia-inducible factor 1 (HIF-1) signaling, and steroid hormone biosynthesis pathways. Malathion significantly increased the gene and protein expression of HIF-1α by upregulating key genes in the mitogen-activated protein kinase (MAPK) pathway (Map2k2, Mapk3, and Eif4e2) and the phosphatidylinositol 3-kinase (PI3K) pathway (Pik3r2 and Akt1). Furthermore, malathion downregulated HIF-1α degradation-regulating genes while upregulating anaerobic metabolism and inflammation-related genes, thereby inhibiting normoxia and promoting hypoxia processes. Testicular hypoxia subsequently induced steroid hormone biosynthesis disorders and spermatogenesis dysfunction. Molecular docking verified that malathion interfered with HIF-1α and steroid hormone synthases (CYP11A1, CYP17A1 and CYP19A1) by forming hydrogen bonds and hydrophobic interactions with these proteins. This study presents the first evidence that malathion triggers spermatogenesis dysfunction in mice through activating the HIF-1/MAPK/PI3K pathway, providing a comprehensive understanding of the reproductive toxicity risks associated with organophosphorus pesticides.
Collapse
Affiliation(s)
- Shouchun Xiao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Jingna Cui
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Yue Cao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Yaru Zhang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Jiaxing Yang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Li Zheng
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Fanrong Zhao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Xueke Liu
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Donghui Liu
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Peng Wang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No.2 West Yuanmingyuan Road, Beijing, 100193, PR China.
| |
Collapse
|
21
|
Shi J, Song S, Wang Y, Wu K, Liang G, Wang A, Xu X. Esketamine alleviates ferroptosis-mediated acute lung injury by modulating the HIF-1α/HO-1 pathway. Int Immunopharmacol 2024; 142:113065. [PMID: 39243557 DOI: 10.1016/j.intimp.2024.113065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/07/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Alveolar epithelial cell (AEC) ferroptosis contributes to the progression of acute lung injury (ALI). Esketamine (ESK) is a new clinical sedative, anesthetic, and analgesic drug that has attracted substantial attention in mental health research because of its antidepressant effects. However, the effects of ESK on ferroptosis-mediated ALI remain unclear. OBJECTIVE This study aimed to explore the protective effect of ESK on AEC ferroptosis in ALI and its potential molecular mechanism in vivo and in vitro. METHODS The antiferroptotic and anti-inflammatory effects of ESK were assessed in a mouse model of lipopolysaccharide (LPS)-induced ALI. In vitro, the epithelial cell lines MLE-12 and A549 were used to examine the underlying mechanism by which ESK regulates inflammation and ferroptosis. RESULTS ESK protected mice against LPS-induced ALI, significantly attenuated pathological changes in the lungs and decreased inflammation and ferroptosis. In vitro, ESK inhibited LPS-induced inflammation and ferroptosis in MLE-12 and A549 cells. Moreover, ferroptosis mediated inflammation in LPS-induced ALI in vivo and in vitro, and ESK decreased the LPS-induced inflammatory response by suppressing ferroptosis. ESK promoted the HIF-1α/HO-1 pathway in LPS-treated AECs and in the lung tissues of mice with LPS-induced ALI. Moreover, pretreatment with ESK and the HIF-1α stabilizer dimethyloxaloylglycine (DMOG) substantially attenuated lung injury and prevented changes in ferroptosis-related biochemical indicators, including glutathione (GSH) depletion, malondialdehyde (MDA) production and glutathione peroxidase 4 (GPX4) downregulation, in untreated LPS-induced mice but not in LPS-induced mice treated with the HO-1 inhibitor zinc protoporphyrin (ZNPP). Similar effects were observed in vitro in HO-1 siRNA-transfected A549 cells after LPS incubation but not in control siRNA-transfected cells. CONCLUSION ESK can inhibit ferroptosis-mediated lipid peroxidation by increasing the expression of HIF-1α/HO-1 pathway, highlighting the potential of ESK to treat LPS-induced ALI.
Collapse
Affiliation(s)
- Jinye Shi
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Shuang Song
- Department of Respiratory Medicine, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Yajie Wang
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Kaixuan Wu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Gui Liang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Aizhong Wang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| | - Xiaotao Xu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| |
Collapse
|
22
|
Ni F, Wang F, Sun J, Tu M, Chen J, Shen X, Ye X, Chen R, Liu Y, Sun X, Chen J, Li X, Zhang D. Proteome-wide Mendelian randomization and functional studies uncover therapeutic targets for polycystic ovarian syndrome. Am J Hum Genet 2024; 111:2799-2813. [PMID: 39541979 PMCID: PMC11639085 DOI: 10.1016/j.ajhg.2024.10.008] [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: 08/01/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
Polycystic ovarian syndrome (PCOS) is an endocrine syndrome that affects a large portion of women worldwide. This proteogenomic and functional study aimed to uncover candidate therapeutic targets for PCOS. We comprehensively investigated the causal association between circulating proteins and PCOS using two-sample Mendelian randomization analysis. Cis-protein quantitative trait loci were derived from six genome-wide association studies (GWASs) on plasma proteome. Genetic associations with PCOS were obtained from a large-scale GWAS meta-analysis, FinnGen cohort, and UK Biobank. Colocalization analyses were performed to prioritize the causal role of candidate proteins. Protein-protein interaction (PPI) and druggability evaluation assessed the druggability of candidate proteins. We evaluated the enrichment of tier 1 and 2 candidate proteins in individuals with PCOS and a mouse model and explored the potential application of the identified drug target. Genetically predicted levels of 65 proteins exhibited associations with PCOS risk, with 30 proteins showing elevated levels and 35 proteins showing decreased levels linked to higher susceptibility. PPI analyses revealed that FSHB, POSTN, CCN2, and CXCL11 interacted with targets of current PCOS medications. Eighty medications targeting 20 proteins showed their potential for repurposing as therapeutic targets for PCOS. EGLN1 levels were elevated in granulosa cells and the plasma of individuals with PCOS and in the plasma and ovaries of dehydroepiandrosterone (DHEA)-induced PCOS mouse model. As an EGLN1 inhibitor, administration of roxadustat in the PCOS mouse model elucidated the EGLN1-HIF1α-ferroptosis axis in inducing PCOS and validated its therapeutic effect in PCOS. Our study identifies candidate proteins causally associated with PCOS risk and suggests that targeting EGLN1 provides a promising treatment strategy.
Collapse
Affiliation(s)
- Feida Ni
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China; First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Feixia Wang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Jing Sun
- Department of Big Data in Health Science, School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Mixue Tu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Jianpeng Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Xiling Shen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Xiaohang Ye
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Ruixue Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Yifeng Liu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Xiao Sun
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Jianhua Chen
- Department of Pathology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Xue Li
- Department of Big Data in Health Science, School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China; Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
23
|
Zhang Z, Chen H, Li Q. High-fat diet led to testicular inflammation and ferroptosis via dysbiosis of gut microbes. Int Immunopharmacol 2024; 142:113235. [PMID: 39332089 DOI: 10.1016/j.intimp.2024.113235] [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: 08/13/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
The disorder of gut microbiota has negative impact on male reproductive, and testicular damage is associated with obesity. However, the detailed mechanism of gut microbiota on the obesity-induced testis injury are still unknown. Therefore, we constructed a mouse model to investigate the effects of obesity on testis injury. In this study, we found that HFD-induced obesity could disorder gut microbiota homeostasis, which increased the abundance of Brevundimonas, Desulfovibrionaceae_unclassified and Ralstonia, ultimately leading to the overproduction of lipopolysaccharides (LPS). Meanwhile, HFD-feeding promoted intestinal permeability via inhibiting expression of tight junction proteins (ZO-1, Occludin and Claudin) and reducing excretion of mucus, leading to translocation of LPS. The over-accumulation of LPS in the bloodstream triggered an inflammatory response by activating TLR4/NF-κB pathway in testis. On the other hand, the gut microbiota produced-LPS also could induce ferroptosis in testis, as reflected by enhancing iron content and lipid peroxidation (MDA), as well as decreasing ferroptosis-related proteins, including GPX4, FTH1 and SLC1A11. Moreover, inhibition of LPS ligand (TLR4) with Resatorvid (TAK-242) alleviated obesity-induced testis injury through suppression of inflammation and ferroptosis. In conclusion, this study provides novel insights into the underlying mechanisms of obesity-related testis injury induced by gut microbiota disorder via the gut-testis axis, thus offering potential targets to counteract obesity-induced male reproductive disorder.
Collapse
Affiliation(s)
- Zelin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Huali Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Qingwang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
| |
Collapse
|
24
|
Lin L, Liao Z, Li Y, Pan S, Wu S, Sun QX, Li C. Transcriptomic analysis and validation study of key genes and the HIF‑1α/HO‑1 pathway associated with ferroptosis in neutrophilic asthma. Exp Ther Med 2024; 28:433. [PMID: 39347495 PMCID: PMC11425779 DOI: 10.3892/etm.2024.12722] [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: 02/04/2024] [Accepted: 06/19/2024] [Indexed: 10/01/2024] Open
Abstract
Ferroptosis, as a unique form of cell death caused by iron overload and lipid peroxidation, is involved in the pathogenesis of various inflammatory diseases of the airways. Inhibition of ferroptosis has become a novel strategy for reducing airway epithelial cell death and improving airway inflammation. The aim of the present study was to analyze and validate the key genes and signaling pathways associated with ferroptosis by bioinformatic methods combined with experimental analyzes in vitro and in vivo to aid the diagnosis and treatment of neutrophilic asthma. A total of 1,639 differentially expressed genes (DEGs) were identified in the transcriptome dataset. After overlapping with ferroptosis-related genes, 11 differentially expressed ferroptosis-related genes (DE-FRGs) were obtained. A new diagnostic model was constructed by these DE-FRGs from the transcriptome dataset with those from the GSE108417 dataset. The receiver operating characteristic curve analysis indicated that the area under the curve had good diagnostic performance (>0.8). As a result, four key DE-FRGs (CXCL2, HMOX1, IL-6 and SLC7A5) and biological pathway [hypoxia-inducible factor 1 (HIF-1) signaling pathway] associated with ferroptosis in neutrophilic asthma were identified by the bioinformatics analysis combined with experimental validation. The upstream regulatory network of key DE-FRGs and target drugs were predicted and the molecular docking results from screened 37 potential therapeutic drugs revealed that the 13 small-molecule drugs exhibited a higher stable binding to the primary proteins of key DE-FRGs. The results suggested that four key DE-FRGs and the HIF-1α/heme oxygenase 1 pathway associated with ferroptosis have potential as novel markers or targets for the diagnosis or treatment of neutrophilic asthma.
Collapse
Affiliation(s)
- Lu Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Zenghua Liao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yinghua Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Shitong Pan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Sihui Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qi-Xiang Sun
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chaoqian Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
25
|
Lu C, Zhang Z, Fan Y, Wang X, Qian J, Bian Z. Shikonin induces ferroptosis in osteosarcomas through the mitochondrial ROS-regulated HIF-1α/HO-1 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156139. [PMID: 39423479 DOI: 10.1016/j.phymed.2024.156139] [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: 05/29/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The most common malignant bone tumour is osteosarcoma, which has an unsatisfactory prognosis and unsatisfactory treatment. Ferroptosis shows promise as an effective OS therapy. A substance extracted from the Lithospermum erythrorhizon, Shikonin (SHK), inhibits a number of tumours, including ovarian, gastric, and lung cancers. However, whether SHK induces OS ferroptosis and its mechanisms are not clear. PURPOSE Our study is aimed at investigating whether SHK causes ferroptosis and elucidating its molecular mechanism. METHODS Cell counting Kit-8, cell cycle and cell apoptosis assay were utilised to assess therapeutic effect of SHK against OS. Normal cells, including H9C2, AML-12 and HK-2, haematoxylin and eosin staining and mice serum biomarker tests were used to assess SHK biosafety. Malondialdehyde (MDA) levels, the glutathione (GSH)/oxidized glutathione (GSSG) ratio, reactive oxygen species (ROS), lipid peroxide (LPO), and intracellular Fe2+ detection, quantitative reverse transcription PCR (qRT-PCR), Western blotting (WB) and rescue experiments were employed to confirm whether SHK induced ferroptosis in OS cells. Molecular docking, cellular thermal shift assay (CETSA), and drug affinity responsive target stability (DARTS) assay were used to evaluate the direct binding between SHK and hypoxia-inducible factor-1α (HIF-1α). Protein stability and degradation analysis, small RNA interference, flow cytometry, qRT-PCR, and WB were used to investigate the molecular mechanism of ferroptosis. In vivo xenografts of nude mouse was used to study the anti-OS effect of SHK. RESULTS SHK significantly reduced OS cell viability and induced apoptosis and G2/M arrest. SHK increased intracellular levels of MDA, ROS, LPO, and Fe2+ while simultaneously reducing the GSH/GSSG ratio and GPX4 and SLC7A11 expression. CETSA and DARTS results demonstrated that SHK did not bind targetly to HIF-1α. Instead, mitochondrial ROS (MitoROS) promoted HIF-1α expression, resulting in HO-1 overexpression, excess Fe2+ production, ROS accumulation and GPX depletion, and ferroptosis. Furthermore, inhibition of MitoROS using Mito-TEMPO downregulated HIF-1α/HO-1 axis and mitigated the SHK-induced ferroptosis. In vivo, SHK effectively suppressed OS growth with favourable biosafety, confirming the molecular mechanism underlying SHK-induced ferroptosis in OS. CONCLUSION We observe that HIF-1α/HO-1 axis is the crucial factor in SHK-induced OS ferroptosis. Importantly, we demonstrate that HIF-1α is indirectly regulated by MitoROS rather than SHK bound directly to HIF-1α. Our research suggest that SHK is a potential drug candidate for OS treatment and may help in identifying novel therapeutic targets for OS.
Collapse
Affiliation(s)
- Congcong Lu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, PR China; Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China
| | - Zhen Zhang
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China
| | - Yuhao Fan
- Zhejiang University School of Medicine, Hangzhou, 310000, PR China
| | - Xiyu Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, PR China; Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China
| | - Jin Qian
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China.
| | - Zhenyu Bian
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China.
| |
Collapse
|
26
|
Liu YY, Bao DQ, Zhang ZS, Zhu Y, Liu LM, Li T. Radix Sanguisorbae Improves Intestinal Barrier in Septic Rats via HIF-1 α/HO-1/Fe 2+ Axis. Chin J Integr Med 2024; 30:1101-1112. [PMID: 38212494 DOI: 10.1007/s11655-023-3550-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVE To investigate whether Radix Sanguisorbae (RS, Diyu) could restore intestinal barrier function following sepsis using a cecal ligation and puncture (CLP)-induced septic rat model and lipopolysaccharide (LPS)-challenged IEC-6 cell model, respectively. METHODS Totally 224 rats were divided into 4 groups including a control, sham, CLP and RS group according to a random number table. The rats in the control group were administrated with Ringer's lactate solution (30 mL/kg) with additional dopamine [10 µ g/(kg·min)] and given intramuscular injections of cefuroxime sodium (10 mg/kg) 12 h following CLP. The rats in the RS group were administrated with RS (10 mg/kg) through tail vein 1 h before CLP and treated with RS (10 mg/kg) 12 h following CLP. The rats in the sham group were only performed abdominal surgery without CLP. The rats in the CLP group were performed with CLP without any treatment. The other steps were same as control group. The effects of RS on intestinal barrier function, mesenteric microvessels barrier function, multi-organ function indicators, inflammatory response and 72 h survival window following sepsis were observed. In vitro, the effects of RS on LPS-challenged IEC-6 cell viability, the expressions of zona occludens-1 (ZO-1) and ferroptosis index were evaluated by cell counting kit-8, immunofluorescence and Western blot analysis. Bioinformatic tools were applied to investigate the pharmacological network of RS in sepsis to predict the active compounds and potential protein targets and pathways. RESULTS The sepsis caused severe intestinal barrier dysfunction, multi-organ injury, lipid peroxidation accumulation, and ferroptosis in vivo. RS treatment significantly prolonged the survival time to 56 h and increased 72-h survival rate to 7/16 (43.75%). RS also improved intestinal barrier function and relieved intestinal inflammation. Moreover, RS significantly decreased lipid peroxidation and inhibited ferroptosis (P<0.05 or P<0.01). Administration of RS significantly worked better than Ringer's solution used alone. Using network pharmacology prediction, we found that ferroptosis and hypoxia inducible factor-1 (HIF-1 α) signaling pathways might be involved in RS effects on sepsis. Subsequent Western blot, ferrous iron measurements, and FerroOrange fluorescence of ferrous iron verified the network pharmacology predictions. CONCLUSION RS improved the intestinal barrier function and alleviated intestinal injury by inhibiting ferroptosis, which was related in part to HIF-1 α/heme oxygenase-1/Fe2+ axis.
Collapse
Affiliation(s)
- Yi-Yan Liu
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China
| | - Dai-Qin Bao
- Department of Anesthesiology, Army Medical Center of PLA, Chongqing, 400042, China
| | - Zi-Sen Zhang
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China
| | - Liang-Ming Liu
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China.
| |
Collapse
|
27
|
Wang J, Wei Y, Wu Y, Zhao T, Kang L, Han L, Chen J, Long C, Wei G, Shen L, Wu S. Di-(2-ethylhexyl) phthalate induces prepubertal testicular injury through MAM-related mitochondrial calcium overload in Leydig and Sertoli cell apoptosis. Toxicology 2024; 509:153956. [PMID: 39307383 DOI: 10.1016/j.tox.2024.153956] [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: 06/23/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
As one of the most prevalent environmental endocrine disruptors, di-(2-ethylhexyl) phthalate (DEHP) is known for its significant developmental toxicity to the male reproductive system in humans and mice. Prepubertal exposure to DEHP has been shown to cause testicular damage, but the underlying mechanisms require further investigation. To investigate this effect, prepubertal mice were exposed to 100, 250 or 500 mg/kg body weight (bw) of DEHP for 14 days, which resulted in impaired histological structure and increased apoptosis of the testes. RNA sequencing (RNA-seq) of testicular tissue suggested that DEHP led to injury in Leydig and Sertoli cells. To further elucidate these mechanisms, we conducted experiments using immature mouse Leydig (TM3) and Sertoli (TM4) cells, and exposed them to 200 μM mono-(2-ethylhexyl) phthalate (MEHP), the primary metabolite of DEHP, for 24 h. We found that MEHP exposure induced oxidative stress injury and promoted cell apoptosis, and that cotreatment with N-acetylcysteine partially reversed these injuries. Given the close association between oxidative stress and mitochondrial calcium levels, we demonstrated that MEHP exposure disrupted mitochondria and increased mitochondrial calcium levels. In addition, MEHP exposure facilitated the formation of mitochondria-associated endoplasmic reticulum membranes (MAMs), upregulated protein expression and enhanced the interactions of the IP3R3-Grp75-VDAC1 complex. Furthermore, inhibition of calcium transfer in the IP3R3-Grp75-VDAC1-MCU axis relieved MEHP-induced mitochondrial injury, oxidative stress and apoptosis in TM3 and TM4 cells. This study highlights the importance of MAM-mediated mitochondrial calcium overload and the subsequent apoptosis of Leydig and Sertoli cells as pivotal factors contributing to testicular injury induced by prepubertal exposure to DEHP.
Collapse
Affiliation(s)
- Junke Wang
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuexin Wei
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuhao Wu
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Tianxin Zhao
- Department of Pediatric Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Lian Kang
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lindong Han
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jiadong Chen
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chunlan Long
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Guanghui Wei
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Lianju Shen
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Shengde Wu
- Department of Urology, Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
28
|
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
|
29
|
Li Z, Gan H, Li S, Xue Y, Luo K, Huang K, Zhang Y, Wang Y, Jiang L, Zhang H. Bioinformatics Identification and Validation of Ferroptosis-Related Key Genes and Therapeutic Compounds in Septic Lung Injury. J Inflamm Res 2024; 17:9215-9230. [PMID: 39600675 PMCID: PMC11589777 DOI: 10.2147/jir.s476522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Background Septic lung injury (SLI) is a severe condition with high mortality, and ferroptosis, a form of programmed cell death, is implicated in its pathogenesis. However, the explicit mechanisms underlying this condition remain unclear. This study aimed to elucidate and validate key ferroptosis-related genes involved in the pathogenesis of SLI through bioinformatics analysis and experimental validation. Methods Microarray data related to SLI from the GSE130936 dataset were downloaded from the Gene Expression Omnibus (GEO) database. These data were then intersected with the FerrDb database to obtain ferroptosis-related differentially expressed genes (DEGs). Protein-protein interaction (PPI) networks and functional enrichment analysis were employed to identify key ferroptosis-related DEGs. The Connectivity Map (c-MAP) tool was used to search for potential compounds or drugs that may inhibit ferroptosis-related DEGs. The transcriptional levels of the key genes and potential therapeutic compounds were verified in an LPS-induced mouse model of lung injury. The expression of these key genes was further verified using the GSE60088 and GSE137342 datasets. Results 38 ferroptosis-related DEGs were identified between the septic and control mice. PPI network analysis revealed four modules, the most significant of which included eight ferroptosis-related DEGs. Functional enrichment analysis showed that these genes were enriched in the HIF-1 signaling pathway, including IL-6 (Interleukin-6), TIMP1 (Tissue Inhibitor of Metalloproteinase 1), HIF-1α (Hypoxia-Inducible Factor-1α), and HMOX1 (Heme Oxygenase-1). Phloretin, a natural compound, was identified as a potential inhibitor of these genes. Treatment with phloretin significantly reduced the expression of these genes (p < 0.05), mitigated lung injury, improved inflammatory profiles by approximately 50%, and ferroptosis profiles by nearly 30% in the SLI models. Conclusion This study elucidates the significant role of ferroptosis in SLI and identifies phloretin as a potential therapeutic agent. However, further research, particularly involving human clinical trials, is necessary to validate these findings for clinical use.
Collapse
Affiliation(s)
- Zhile Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Han Gan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Siyuan Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yuchen Xue
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kai Luo
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kai Huang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yunqian Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Hui Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
30
|
Liu J, Ren J, Zhou L, Tan K, Du D, Xu L, Cao W, Zhang Y. Proteomic and lipidomic analysis of the mechanism underlying astragaloside IV in mitigating ferroptosis through hypoxia-inducible factor 1α/heme oxygenase 1 pathway in renal tubular epithelial cells in diabetic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118517. [PMID: 38972525 DOI: 10.1016/j.jep.2024.118517] [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: 04/05/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The limitations of modern medicine in mitigating the pathological process of diabetic kidney disease (DKD) necessitate novel, precise, and effective prevention and treatment methods. Huangqi, the root of Astragalus membranaceus Fisch. ex Bunge has been used in traditional Chinese medicine for various kidney ailments. Astragaloside IV (AS-IV), the primary pharmacologically active compound in A. membranaceus, is involved in lipid metabolism regulation; however, its potential in ameliorating renal damage in DKD remains unexplored. AIM OF THE STUDY To elucidate the specific mechanism by which AS-IV moderates DKD progression. MATERIALS AND METHODS A murine model of DKD and high glucose-induced HK-2 cells were treated with AS-IV. Furthermore, multiomics analysis, molecular docking, and molecular dynamics simulations were performed to elucidate the mechanism of action of AS-IV in DKD, which was validated using molecular biological methods. RESULTS AS-IV regulated glucose and lipid metabolism in DKD, thereby mitigating lipid deposition in the kidneys. Proteomic analysis identified 12 proteins associated with lipid metabolism regulated by AS-IV in the DKD renal tissue. Additionally, lipid metabolomic analysis revealed that AS-IV upregulated and downregulated 4 beneficial and 79 harmful lipid metabolites, respectively. Multiomics analysis further indicated a positive correlation between the top-ranked differential protein heme oxygenase (HMOX)1 and the levels of various harmful lipid metabolites and a negative correlation with the levels of beneficial lipid metabolites. Furthermore, enrichment of both ferroptosis and hypoxia-inducible factor (HIF)-1 signaling pathways during the AS-IV treatment of DKD was observed using proteomic analysis. Validation results showed that AS-IV effectively reduced ferroptosis in DKD-affected renal tubular epithelial cells by inhibiting HIF-1α/HMOX1 pathway activity, upregulating glutathione peroxidase-4 and ferritin heavy chain-1 expression, and downregulating acyl-CoA synthetase long-chain family member-4 and transferrin receptor-1 expression. Our findings demonstrate the potential of AS-IV in mitigating DKD pathology by downregulating the HIF-1α/HMOX1 signaling pathway, thereby averting ferroptosis in renal tubular epithelial cells. CONCLUSIONS AS-IV is a promising treatment strategy for DKD via the inhibition of ferroptosis in renal tubular epithelial cells. The findings of this study may help facilitate the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Jun Liu
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1., Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, PR China.
| | - Jing Ren
- College of Traditional Chinese Medicine, Chongqing Medical and Pharmaceutical College, No. 82, University Town Middle Road, Shapingba District, Chongqing, 401331, PR China.
| | - Linlan Zhou
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1., Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, PR China.
| | - Kaiyue Tan
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1, Medical College Road, Yuzhong District, Chongqing, 400016, PR China; Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, 400016, PR China.
| | - Donglin Du
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, PR China.
| | - Lei Xu
- Laboratory Animal Center, Chongqing Medical University, No. 1, Medical College Road, Yuzhong District, Chongqing, 400016, PR China.
| | - Wenfu Cao
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1., Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, PR China; College of Traditional Chinese Medicine, Chongqing Medical University, No. 1, Medical College Road, Yuzhong District, Chongqing, 400016, PR China; Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, 400016, PR China.
| | - Yudi Zhang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1, Medical College Road, Yuzhong District, Chongqing, 400016, PR China; Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, 400016, PR China; College of Combination of Chinese and Western Medicine, Chongqing College of Traditional Chinese Medicine, No. 61, Puguobao Road, Bicheng Street, Bishan District, Chongqing, 402760, PR China.
| |
Collapse
|
31
|
Yang H, Ding L, Xu B, Zhang Z, Dai W, He T, Liu L, Du X, Fu X. Lycium barbarum polysaccharide alleviates ferroptosis in Sertoli cells through NRF2/SLC7A11/GPX4 pathway and ameliorates DEHP-induced male reproductive damage in mice. Int J Biol Macromol 2024; 282:137241. [PMID: 39515713 DOI: 10.1016/j.ijbiomac.2024.137241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/24/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Di-(2-ethylhexyl)phthalate (DEHP) is a common plasticizer that has been shown to significantly negatively affect male reproductive health. On the other hand, Lycium barbarum polysaccharide (LBP) has been shown to improve reproductive function. Therefore, we hypothesized that LBP may ameliorate DEHP-induced male reproductive damage. Herein, we found that LBP could alleviate DEHP-induced testicular damage and sperm abnormalities. Furthermore, histomorphological analysis of mice testis revealed that LBP primarily ameliorated the DEHP-induced male reproductive damage by targeting Sertoli cells. Moreover, the detection of the function-related genes of Sertoli cells confirmed this finding. The serum of mice in the Control, DEHP, and DEHP+LBP groups was analyzed using non-targeted metabolomics to further elucidate the mechanism of action of LBP in improving DEHP-induced male reproductive damage. According to the results, the differential metabolites were mainly enriched in the glutamate metabolism pathway, implying that LBP may alleviate the ferroptosis-related DEHP-induced testicular injury. Related ferroptosis markers were also found in mice testis. These findings collectively suggest that LBP may ameliorate DEHP-induced testicular injury via alleviating ferroptosis in Sertoli cells. To clarify the specific mechanism, we constructed a cell model in vitro by treating TM4 cells (the Sertoli cell line) with LBP and MEHP (the in vivo DEHP metabolite). Our findings revealed that LBP can improve the function of DEHP-affected Sertoli cells. Furthermore, the analysis of lipid peroxidation, Fe2+ content, and ferroptosis-related protein expressions demonstrated that LBP could ameliorate MEHP-induced ferroptosis in TM4 cells. To clarify the specific mechanism, glutamate metabolism-related proteins involved in the ferroptosis pathway were detected. According to the results, there were significant changes in the expression of NRF2, SLC7A11 and GPX4 proteins, which are involved in the ferroptosis glutamate metabolism pathway. Furthermore, supplementation of NRF2, SLC7A11, and GPX4 inhibitors (ML385, Erastin, and RSL3, respectively) blocked the therapeutic effect of LBP in alleviating MEHP-induced ferroptosis in TM4 cells, implying that LBP could also ameliorate MEHP-induced ferroptosis via the NRF2/SLC7A11/GPX4 pathway. In summary, these findings show that LBP can alleviate DEHP/MEHP-induced ferroptosis through the NRF2/SLC7A11/GPX4 pathway, ameliorating Sertoli cell dysfunction and improving the DEHP-induced male reproductive damage. Therefore, the clinical administration of LBP could be an effective strategy for preventing DEHP-induced male reproductive injury.
Collapse
Affiliation(s)
- Hong Yang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Liyang Ding
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Bo Xu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Zhen Zhang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Wenjie Dai
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Tiantian He
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Ling Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xing Du
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China.
| | - Xufeng Fu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
32
|
Xu X, He Y, Cheng Z, Zhang H, Chu Y, Wang Z, An X. Environmental endocrine disrupting chemical-DEHP exposure-provoked biotoxicity about microbiota-gut-mammary axis in lactating mice via multi-omics technologies. ENVIRONMENT INTERNATIONAL 2024; 193:109130. [PMID: 39522489 DOI: 10.1016/j.envint.2024.109130] [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: 04/12/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Plastics, pervasive in humans and nature, often contain Di (2-ethylhexyl) phthalate (DEHP) that enhance plastic's elasticity. However, DEHP is an environmental endocrine disruptor, affecting organisms upon exposure. Understanding mammary gland development in lactating females is crucial for offspring nourishment and dairy production. Employing multi-omics technology, this study aimed to uncover DEHP's impact on the microbial-gut-mammary axis. Forty mice were exposed to varying DEHP doses for 18 d. We performed 16S sequencing, metabolomics, mammary tissue observation, and gene expression profiling. Results revealed DEHP's influence on microbial diversity, with increased Lactobacillus abundance and reduced Proteobacteria, alongside colonic inflammation. Elevated GMP and adenosine 5'-monophosphate levels in the bloodstream were noted, while ascorbic acid, glycitein, and others decreased. MEHP, a DEHP metabolite, damaged mammary tissues, inhibiting ERK1/2 phosphorylation, triggering apoptosis and ferroptosis. These findings unveil potential therapeutic targets for DEHP-induced chronic toxicity in humans and animals, aiding dairy livestock health and human well-being. This study underscores the importance of understanding the adverse effects of DEHP exposure on mammalian systems.
Collapse
Affiliation(s)
- Xiaolong Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yonglong He
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zefang Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Haoyuan Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yijian Chu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhewei Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
33
|
Huang X, Fu Y, Wang S, Guo Q, Wu Y, Zheng X, Wang J, Wu S, Shen L, Wei G. 2,2',4,4'-Tetrabromodiphenyl ether exposure disrupts blood-testis barrier integrity through CMA-mediated ferroptosis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 948:174738. [PMID: 39009145 DOI: 10.1016/j.scitotenv.2024.174738] [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: 05/29/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
2,2',4,4'-Tetrabromodiphenyl ether (PBDE-47), being the most prevalent congener of polybrominated diphenyl ethers (PBDEs), has been found to accumulate greatly in the environment and induce spermatogenesis dysfunction. However, the specific underlying factors and mechanisms have not been elucidated. Herein, male Sprague-Dawley (SD) rats were exposed to corn oil, 10 mg/kg body weight (bw) PBDE-47 or 20 mg/kg bw PBDE-47 by gavage for 30 days. PBDE-47 exposure led to blood-testis barrier (BTB) integrity disruption and aberrant spermatogenesis. Given that Sertoli cells are the main toxicant target, to explore the potential mechanism involved, we performed RNA sequencing (RNA-seq) in Sertoli cells, and the differentially expressed genes were shown to be enriched in ferroptosis and lysosomal pathways. We subsequently demonstrated that ferroptosis was obviously increased in testes and Sertoli cells upon exposure to PBDE-47, and the junctional function of Sertoli cells was restored after treatment with the ferroptosis inhibitor ferrostatin-1. Since glutathione peroxidase 4 (GPX4) was dramatically reduced in PBDE-47-exposed testes and Sertoli cells and considering the RNA-sequencing results, we examined the activity of chaperone-mediated autophagy (CMA) and verified that the expression of LAMP2a and HSC70 was upregulated significantly after PBDE-47 exposure. Notably, Lamp2a knockdown not only inhibited ferroptosis by suppressing GPX4 degradation but also restored the impaired junctional function induced by PBDE-47. These collective findings strongly indicate that PBDE-47 induces Sertoli cell ferroptosis through CMA-mediated GPX4 degradation, resulting in decreased BTB-associated protein expression and eventually leading to BTB integrity disruption and spermatogenesis dysfunction.
Collapse
Affiliation(s)
- Xu Huang
- Department of Urology, Children's Hospital of Chongqing Medical University, China; Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, China
| | - Yan Fu
- Department of Urology, Children's Hospital of Chongqing Medical University, China; Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, China
| | - Siyuan Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, China; Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, China
| | - Qitong Guo
- Department of Urology, Children's Hospital of Chongqing Medical University, China; Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, China
| | - Yuhao Wu
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangqin Zheng
- Department of Urology, Children's Hospital of Chongqing Medical University, China; Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, China
| | - Junke Wang
- Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, China; Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, China
| | - Lianju Shen
- Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, China.
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, China; Pediatric Research Institute, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, China.
| |
Collapse
|
34
|
Shi Y, Yin L, Li JY, Zhou SM, Wang N, Chen HQ, Zeng Y, Li YW, Liu WB. FTO mediates bisphenol F-induced blood-testis barrier impairment through regulating ferroptosis via YTHDF1/TfRc and YTHDF2/SLC7A11 signal axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 359:124531. [PMID: 38996995 DOI: 10.1016/j.envpol.2024.124531] [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: 05/08/2024] [Revised: 06/30/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Bisphenol F (BPF) has been extensively utilized in daily life, which brings new hazards to male reproductive health. However, the specific functional mechanism is still unclear. Both cell and animal models were utilized for exploring the role of RNA methylation and ferroptosis and its underlying mechanisms in male reproductive injury induced by BPF. In animal model, BPF severely destroyed the integrity of the blood-testis barrier (BTB) and induced ferroptosis. Furthermore, BPF significantly affected the barrier function of TM4 cells and promoted ferroptosis. Importantly, ChIP assays revealed that BPF inhibited AR transcriptional regulation of FTO and FTO expression was downregulated in TM4 cells. Overexpression of FTO prevented the impairment of BTB by inhibiting ferroptosis in TM4 cells. Mechanistically, FTO could significantly down-regulate the m6A modification level of TfRc and SLC7A11 mRNA through MeRIP experiment. RIP experiments showed that YTHDF1 can bind to TfRc mRNA and promote its translation while YTHDF2 could bind to SLC7A11 mRNA and reduce its mRNA stability. Therefore, our results suggest that FTO plays a key role in BPF induced male reproductive toxicity through YTHDF1-TfRc axis and YTHDF2-SLC7A11 axis and may provide new ideas and methods for the prevention and treatment of male reproductive diseases associated with environmental pollutants.
Collapse
Affiliation(s)
- Yu Shi
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China; Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Li Yin
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jiang-Ying Li
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China; Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shi-Meng Zhou
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Na Wang
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Hong-Qiang Chen
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yong Zeng
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ya-Wen Li
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Wen-Bin Liu
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
35
|
Meng J, Xiao L, Li Q, Gong L, Luo P, Zhao Y, Wang S. Di-(2-ethylhexyl) phthalate exposure induces ferroptosis by regulating the Nrf2-mediated signaling pathway in mouse ovaries. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117104. [PMID: 39321527 DOI: 10.1016/j.ecoenv.2024.117104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/19/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), an endocrine-disrupting chemical present in plasticized products, exerts strong modulation on the anatomy and function of the female reproductive system. However, the potential mechanisms underlying DEHP-induced regulation of prepubertal female reproductive toxicity have not yet been elucidated. Therefore, this study was designed to elucidate the molecular mechanism of ovarian injury induced by DEHP exposure in mice. Elevated serum mono-2-ethylhexyl phthalate (MEHP) concentrations, decreased levels of ovarian hormones (E2 and P4), and observed ovarian injury were found after DEHP exposure. Ovarian transcriptome analysis revealed significant alterations in ferroptosis-associated gene expression, with potential regulation by Nrf2. Subsequent analysis of ferrous iron, malondialdehyde (MDA), Western blotting, and immunofluorescence of the ovaries confirmed the RNA-seq findings. Transcriptome analysis of granulosa cells revealed a direct or indirect regulatory relationship between Nrf2 and downstream ferroptosis-related proteins following MEHP exposure. Further experiments demonstrated that ferrostatin-1 attenuated MEHP-induced ferroptosis in granulosa cells. Additionally, Nrf2 stabilization and accumulation in the nucleus of granulosa cells were observed following MEHP treatment. RNAi-mediated knockdown of Nrf2 exacerbated MEHP-induced ferroptosis in granulosa cells. This evidence indicates that DEHP exposure induces ferroptosis through regulation of the Nrf2-mediated signaling pathway in mouse ovaries, laying the groundwork for future studies aiming to develop therapeutic strategies against DEHP toxicity.
Collapse
Affiliation(s)
- Jinzhu Meng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China; Guizhou Provincial Key Laboratory for Biodiversity Conservation and Utilization in the Fanjing Mountain Region, Tongren University, Tongren, China
| | - Lilin Xiao
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Qiuye Li
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Ling Gong
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Ping Luo
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Yuanyuan Zhao
- Guizhou Provincial Key Laboratory for Biodiversity Conservation and Utilization in the Fanjing Mountain Region, Tongren University, Tongren, China.
| | - Shuilian Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.
| |
Collapse
|
36
|
Lu Z, Huang Q, Qin X, Chen F, Li E, Lin H. Novel Insights into Ethanol-Soluble Oyster Peptide-Zinc-Chelating Agents: Structural Characterization, Chelation Mechanism, and Potential Protection on MEHP-Induced Leydig Cells. Mar Drugs 2024; 22:465. [PMID: 39452873 PMCID: PMC11509544 DOI: 10.3390/md22100465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Numerous studies have reported that mono-(2-ethylhexyl) phthalate (MEHP) (bioactive metabolite of Di(2-ethylhexyl) phthalate) has inhibitory effects on Leydig cells. This study aims to prepare an oyster peptide-zinc complex (PEP-Zn) to alleviate MEHP-induced damage in Leydig cells. Zinc-binding peptides were obtained through the following processes: zinc-immobilized affinity chromatography (IMAC-Zn2+), liquid chromatography-mass spectrometry technology (LC-MS/MS) analysis, molecular docking, molecular dynamic simulation, and structural characterization. Then, the Zn-binding peptide (PEP) named Glu-His-Ala-Pro-Asn-His-Asp-Asn-Pro-Gly-Asp-Leu (EHAPNHDNPGDL) was identified. EHAPNHDNPGDL showed the highest zinc-chelating ability of 49.74 ± 1.44%, which was higher than that of the ethanol-soluble oyster peptides (27.50 ± 0.41%). In the EHAPNHDNPGDL-Zn complex, Asn-5, Asp-7, Asn-8, His-2, and Asp-11 played an important role in binding to the zinc ion. Additionally, EHAPNHDNPGDL-Zn was found to increase the cell viability, significantly increase the relative activity of antioxidant enzymes and testosterone content, and decrease malondialdehyde (MDA) content in MEHP-induced TM3 cells. The results also indicated that EHAPNHDNPGDL-Zn could alleviate MEHP-induced apoptosis by reducing the protein level of p53, p21, and Bax, and increasing the protein level of Bcl-2. These results indicate that the zinc-chelating peptides derived from oyster peptides could be used as a potential dietary zinc supplement.
Collapse
Affiliation(s)
- Zhen Lu
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, College of Food Science and Technology, National Research and Development Branch Center for Shellfish Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Ocean University, Zhanjiang 524088, China; (Z.L.); (Q.H.)
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian 463000, China; (F.C.); (E.L.)
| | - Qianqian Huang
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, College of Food Science and Technology, National Research and Development Branch Center for Shellfish Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Ocean University, Zhanjiang 524088, China; (Z.L.); (Q.H.)
| | - Xiaoming Qin
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, College of Food Science and Technology, National Research and Development Branch Center for Shellfish Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Ocean University, Zhanjiang 524088, China; (Z.L.); (Q.H.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Fujia Chen
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian 463000, China; (F.C.); (E.L.)
| | - Enzhong Li
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian 463000, China; (F.C.); (E.L.)
| | - Haisheng Lin
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, College of Food Science and Technology, National Research and Development Branch Center for Shellfish Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Ocean University, Zhanjiang 524088, China; (Z.L.); (Q.H.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
37
|
Guo Z, Zhang W, Gao H, Li Y, Li X, Yang X, Fan L. High expression levels of haem oxygenase-1 promote ferroptosis in macrophage-derived foam cells and exacerbate plaque instability. Redox Biol 2024; 76:103345. [PMID: 39255694 PMCID: PMC11414708 DOI: 10.1016/j.redox.2024.103345] [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: 08/08/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
Plaque rupture with consequent thrombosis is the leading cause of acute cardiovascular events, during which macrophage death is a hallmark. Ferroptosis is a pivotal intermediate link between early and advanced atherosclerosis. Existing evidence indicates the involvement of macrophage ferroptosis in plaque vulnerability; however, the exact mechanism remains elusive. The aim of this study was to explore key ferroptosis-related genes (FRGs) involved in plaque progression and the underlying molecular mechanisms involved. The expression landscape of FRGs was obtained from atherosclerosis-related GEO datasets. Molecular mechanism studies of ferroptosis were performed using bone marrow-derived macrophages (BMDMs) and macrophage-derived foam cells (MDFCs). Bioinformatics analysis and immunohistochemistry revealed that macrophage haem oxygenase-1 (HMOX1) is the key FRG involved in plaque destabilization. Hypoxic conditions induced a significant increase in Hmox1 expression in MDFCs but not in macrophages. In addition, the beneficial or deleterious effects of Hmox1 were dependent on the degree of Hmox1 induction. Hmox1 overexpression drove inflammatory responses and ferroptotic oxidative stress in MDFCs and aggravated the plaque burden in atherosclerotic model mice. Further mechanistic investigations demonstrated that hypoxia-mediated degradation of egl-9 family hypoxia-inducible factor 3 (Egln3) stabilized Hif1a, which subsequently promoted Hmox1 transcription. Our findings suggest that high Hmox1 expression under hypoxia is deleterious to MDFC viability and plaque stability, providing a reference for the management of acute cardiovascular events.
Collapse
Affiliation(s)
- Zhenyu Guo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wan Zhang
- Department of Vascular Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Hongxia Gao
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Yang Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xu Li
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Xiaohu Yang
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| | - Longhua Fan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China.
| |
Collapse
|
38
|
Fan CH, Zeng XQ, Feng RM, Yi HW, Xia R. Comprehensive review of perioperative factors influencing ferroptosis. Biomed Pharmacother 2024; 179:117375. [PMID: 39278186 DOI: 10.1016/j.biopha.2024.117375] [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: 07/17/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
The perioperative period encompasses all phases of patient care from the decision to perform surgery until full recovery. Ferroptosis, a newly identified type of regulated cell death, influences a wide array of diseases, including those affecting the prognosis and regression of surgical patients, such as ischemia-reperfusion injury and perioperative cognitive dysfunction. This review systematically examines perioperative factors impacting ferroptosis such as surgical trauma-induced stress, tissue hypoxia, anesthetics, hypothermia, and blood transfusion. By analyzing their intrinsic relationships, we aim to improve intraoperative management, enhance perioperative safety, prevent complications, and support high-quality postoperative recovery, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Cheng-Hui Fan
- Department of Anaesthesiology, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, PR China
| | - Xiao-Qin Zeng
- Department of Anaesthesiology, The Second People's Hospital of Jingzhou, Jingzhou 434020, PR China
| | - Rui-Min Feng
- Laboratory Department, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, PR China
| | - Hua-Wei Yi
- Laboratory Department, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, PR China.
| | - Rui Xia
- Department of Anaesthesiology, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, PR China.
| |
Collapse
|
39
|
Cao H, Xie Q, Luo P, Chen J, Xia K, Ma L, Chen D, Deng C, Wan Z. Di-(2-ethylhexyl) phthalate exposure induces premature testicular senescence by disrupting mitochondrial respiratory chain through STAT5B-mitoSTAT3 in Leydig cell. GeroScience 2024; 46:4373-4396. [PMID: 38499958 PMCID: PMC11336147 DOI: 10.1007/s11357-024-01119-x] [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: 10/04/2023] [Accepted: 02/27/2024] [Indexed: 03/20/2024] Open
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), a prevalent plasticizer, is known to have endocrine-disrupting effects on males and cause reproductive toxicity. There were causal effects of DEHP on testosterone levels in the real world by Mendelian randomization analysis. Exposure to DEHP during the preadult stage might lead to premature testicular senescence, but the mechanisms responsible for this have yet to be determined. In this study, we administered DEHP (300 mg/kg/day) to male C57BL/6 mice from postnatal days 21 to 49. The mice were kept for 6 months without DEHP. RNA sequencing was conducted on testicular tissue at PNM6. The results indicated that DEHP hindered testicular development, lowered serum testosterone levels in male mice, and induced premature testicular senescence. TM3 Leydig cells were exposed to 300 μM of mono(2-ethylhexyl) phthalate (MEHP), the bioactive metabolite of DEHP, for 72 h. The results also found that DEHP/MEHP induced senescence in vivo and in vitro. The mitochondrial respiratory chain was disrupted in Leydig cells. The expression and stability of STAT5B were elevated by MEHP treatment in TM3 cells. Furthermore, p-ERK1/2 was significantly decreased by STAT5B, and mitochondria-STAT3 (p-STAT3 ser727) was significantly decreased due to the decrease of p-ERK1/2. Additionally, the senescence level of TM3 cells was decreased and treated with 5 mM NAC for 1 h after MEHP treatment. In conclusion, these findings provided a novel mechanistic understanding of Leydig cells by disrupting the mitochondrial respiratory chain through STAT5B-mitoSTAT3.
Collapse
Affiliation(s)
- Haiming Cao
- The Andrology Department, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- The Reproductive Andrology Clinic, the Seventh Affiliated Hospital of Sun Yat-Sen University, 628 Zhenyuan Road, 518000, Shenzhen, Guangdong, China
| | - Qigen Xie
- The Andrology Department, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- The Department of Pediatric Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Peng Luo
- The Andrology Department, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Jiaqi Chen
- The Urology Department, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, 365000, Fujian, China
| | - Kai Xia
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Lin Ma
- The Reproductive Center, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518000, Guangdong, China
| | - Demeng Chen
- Translational Medicine Center, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Chunhua Deng
- The Andrology Department, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Zi Wan
- The Andrology Department, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
40
|
Qian Y, Liu Y, Wang T, Wang S, Chen J, Li F, Zhang M, Hu X, Wang J, Li Y, James A, Hou R, Cai K. Effects of Cryptorchidism on the Semen Quality of Giant Pandas from the Perspective of Seminal Plasma Proteomics. Genes (Basel) 2024; 15:1288. [PMID: 39457412 PMCID: PMC11507308 DOI: 10.3390/genes15101288] [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: 08/17/2024] [Revised: 09/16/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Giant pandas are an endangered species with low reproductive rates. Cryptorchidism, which can negatively affect reproduction, is also often found in pandas. Seminal plasma plays a crucial role in sperm-environment interactions, and its properties are closely linked to conception potential in both natural and assisted reproduction. The research sought to identify seminal fluid protein content variations between normal and cryptorchid giant pandas. Methods: Using a label-free MS-based method, the semen proteomes of one panda with cryptorchidism and three normal pandas were studied, and the identified proteins were compared and functionally analyzed. Results: Mass spectrometry identified 2059 seminal plasma proteins, with 361 differentially expressed proteins (DEPs). Gene ontology (GO) analysis revealed that these DEPs are mainly involved in the phosphate-containing compound metabolic, hydrolase activity, and kinase activity areas (p ≤ 0.05). The KEGG functional enrichment analysis revealed that the top 20 pathways were notably concentrated in the adipocyte lipolysis and insulin metabolism pathway, with a significance level of p ≤ 0.05. Further analysis through a protein-protein interaction (PPI) network identified nine key proteins that may play crucial roles, including D2GXH8 (hexokinase Fragment), D2HSQ6 (protein tyrosine phosphatase), and G1LHZ6 (Calmodulin 2). Conclusions: We suspect that the high abundance of D2HSQ6 in cryptorchid individuals is associated with metabolic pathways, especially the insulin signal pathway, as a typical proteomic feature related to its pathological features. These findings offer insight into the ex situ breeding conditions of this threatened species.
Collapse
Affiliation(s)
- Yicheng Qian
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (Y.Q.); (T.W.)
| | - Yuliang Liu
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Tao Wang
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (Y.Q.); (T.W.)
| | - Shenfei Wang
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Jiasong Chen
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Feiping Li
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Mengshi Zhang
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Xianbiao Hu
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Juan Wang
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Yan Li
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Ayala James
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Rong Hou
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| | - Kailai Cai
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China; (Y.L.); (S.W.); (J.C.); (F.L.); (M.Z.); (X.H.); (J.W.); (Y.L.); (A.J.); (R.H.)
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
| |
Collapse
|
41
|
Cao H, Li Z, Jin T, He S, Liu S, Li L, Wang Y, Gong Y, Wang G, Yang F, Dong W. Maslinic acid supplementation prevents di(2-ethylhexyl) phthalate-induced apoptosis via PRDX6 in peritubular myoid cells of Chinese forest musk deer. J Environ Sci (China) 2024; 143:47-59. [PMID: 38644023 DOI: 10.1016/j.jes.2023.07.034] [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: 03/22/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 04/23/2024]
Abstract
Chinese forest musk deer (FMD), an endangered species, have exhibited low reproductive rates even in captivity due to stress conditions. Investigation revealed the presence of di(2-ethylhexyl) phthalate (DEHP), an environmental endocrine disruptor, in the serum and skin of captive FMDs. Feeding FMDs with maslinic acid (MA) has been observed to alleviate the stress response and improve reproductive rates, although the precise molecular mechanisms remain unclear. Therefore, this study aims to investigate the molecular mechanisms underlying the alleviation of DEHP-induced oxidative stress and cell apoptosis in primary peritubular myoid cells (PMCs) through MA intake. Primary PMCs were isolated and exposed to DEHP in vitro. The results demonstrated that DEHP significantly suppressed antioxidant levels and promoted cell apoptosis in primary PMCs. Moreover, interfering with the expression of PRDX6 was found to induce excessive reactive oxygen species (ROS) production and cell apoptosis in primary PMCs. Supplementation with MA significantly upregulated the expression of PRDX6, thereby attenuating DEHP-induced oxidative stress and cell apoptosis in primary PMCs. These findings provide a theoretical foundation for mitigating stress levels and enhancing reproductive capacity of in captive FMDs.
Collapse
Affiliation(s)
- Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China
| | - Zhenpeng Li
- College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China; Shaanxi Qiyuan-Times Agri-Tech Development Co. Ltd., Shaanxi 725000, China
| | - Tianqi Jin
- College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China; Biology Research Centre of Qin-Mountains Wildlife, Northwest A&F University, Shaanxi 712100, China
| | - Shuyang He
- College of Forestry, Northwest A&F University, Shaanxi 712100, China; Biology Research Centre of Qin-Mountains Wildlife, Northwest A&F University, Shaanxi 712100, China
| | - Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China
| | - Long Li
- College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China; Biology Research Centre of Qin-Mountains Wildlife, Northwest A&F University, Shaanxi 712100, China
| | - Yang Wang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China; Biology Research Centre of Qin-Mountains Wildlife, Northwest A&F University, Shaanxi 712100, China; Shaanxi Qiyuan-Times Agri-Tech Development Co. Ltd., Shaanxi 725000, China
| | - Ye Gong
- College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China; Shaanxi Qiyuan-Times Agri-Tech Development Co. Ltd., Shaanxi 725000, China
| | - Gang Wang
- Shaanxi Qiyuan-Times Agri-Tech Development Co. Ltd., Shaanxi 725000, China
| | - Fangxia Yang
- College of Forestry, Northwest A&F University, Shaanxi 712100, China; Biology Research Centre of Qin-Mountains Wildlife, Northwest A&F University, Shaanxi 712100, China.
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China; Biology Research Centre of Qin-Mountains Wildlife, Northwest A&F University, Shaanxi 712100, China
| |
Collapse
|
42
|
Jiahong C, Junfeng D, Shuxian L, Tao W, Liyun W, Hongfu W. The role of immune cell death in spermatogenesis and male fertility. J Reprod Immunol 2024; 165:104291. [PMID: 38986230 DOI: 10.1016/j.jri.2024.104291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024]
Abstract
The male reproductive system provides a distinctive shield to the immune system, safeguarding germ cells (GCs) from autoimmune harm. The testis in mammals creates a unique immunological setting due to its exceptional immune privilege and potent local innate immunity. which can result from a number of different circumstances, including disorders of the pituitary gland, GC aplasia, and immunological elements. Apoptosis, or programmed cell death (PCD), is essential for mammalian spermatogenesis to maintain and ensure an appropriate number of GCs that correspond with the supporting capability of the Sertoli cells. Apoptosis is substantial in controlling the number of GCs in the testis throughout spermatogenesis, and any dysregulation of this process has been linked to male infertility. There is a number of evidence about the potential of PCD in designing novel therapeutic approaches in the treatment of infertility. A detailed understanding of PCD and the processes that underlie immunological infertility can contribute to the progress in designing strategies to prevent and treat male infertility. This review will provide a summary of the role of immune cell death in male reproduction and infertility and describe the therapeutic strategies and agents for treatment based on immune cell death.
Collapse
Affiliation(s)
- Chen Jiahong
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Department of Venereal Diseases and Integrated Chinese and Western Medicine and Bone Paralysis, Longjiang Hospital of Shunde District, Foshan, China
| | - Dong Junfeng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Liu Shuxian
- Guangzhou Huadu District Maternal and Child Health Care Hospital (Huzhong Hospital of Huadu District), Guangzhou, China
| | - Wang Tao
- Department of Venereal Diseases and Integrated Chinese and Western Medicine and Bone Paralysis, Longjiang Hospital of Shunde District, Foshan, China.
| | - Wang Liyun
- Guangzhou Huadu District Maternal and Child Health Care Hospital (Huzhong Hospital of Huadu District), Guangzhou, China.
| | - Wu Hongfu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
43
|
Sarangi P, Sahoo PK, Pradhan LK, Bhoi S, Sahoo BS, Chauhan NR, Raut S, Das SK. Concerted monoamine oxidase activity following exposure to di-2-ethylhexyl phthalate is associated with aggressive neurobehavioral response and neurodegeneration in zebrafish brain. Comp Biochem Physiol C Toxicol Pharmacol 2024; 283:109970. [PMID: 38944366 DOI: 10.1016/j.cbpc.2024.109970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/01/2024]
Abstract
Di-2-ethylhexyl phthalate (DEHP) is the most commonly preferred synthetic organic chemical in plastics and its products for making them ductile, flexible and durable. As DEHP is not chemically bound to the macromolecular polymer of plastics, it can be easily leached out to accumulate in food and environment. Our recent report advocated that exposure to DEHP significantly transformed the innate bottom-dwelling and scototaxis behaviour of zebrafish. Our present study aimed to understand the possible role of DEHP exposure pertaining towards the development of aggressive behaviour and its association with amplified monoamine oxidase activity and neurodegeneration in the zebrafish brain. As heightened monoamine oxidase (MAO) is linked with genesis of aggressive behaviour, our observation also coincides with DEHP-persuaded aggressive neurobehavioral transformation in zebrafish. Our preliminary findings also showed that DEHP epitomized as a prime factor in transforming native explorative behaviour and genesis of aggressive behaviour through oxidative stress induction and changes in the neuromorphology in the periventricular grey zone (PGZ) of the zebrafish brain. With the finding demarcating towards heightened chromatin condensation in the PGZ of zebrafish brain, our further observation by immunohistochemistry showed a profound augmentation in apoptotic cell death marker cleaved caspase 3 (CC3) expression following exposure to DEHP. Our further observation by immunoblotting study also demarcated a temporal augmentation in CC3 and tyrosine hydroxylase expression in the zebrafish brain. Therefore, the gross findings of the present study delineate the idea that chronic exposure to DEHP is associated with MAO-instigated aggressive neurobehavioral transformation and neurodegeneration in the zebrafish brain.
Collapse
Affiliation(s)
- Prerana Sarangi
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar 751003, India
| | - Pradyumna Kumar Sahoo
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar 751003, India
| | - Lilesh Kumar Pradhan
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar 751003, India; Centre of Excellence, Natural Products and Therapeutics Laboratory, Department of Biotechnology and Bioinformatics, Sambalpur University, Odisha 768019, India
| | - Suvam Bhoi
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar 751003, India
| | - Bhabani Sankar Sahoo
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar 751003, India; Institute of Life Sciences, NALCO Nagar, Chandrasekharpur, Bhubaneswar, Odisha 751023, India
| | - Nishant Ranjan Chauhan
- Department of Neurobiology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Sangeeta Raut
- Environmental Biotechnology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar 751003, India
| | - Saroj Kumar Das
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar 751003, India; Department of Zoology, Kuntala Kumari Sabat Women's College, Balasore, Odisha 756003, India.
| |
Collapse
|
44
|
Lv YT, Liu TB, Li Y, Wang ZY, Lian CY, Wang L. HO-1 activation contributes to cadmium-induced ferroptosis in renal tubular epithelial cells via increasing the labile iron pool and promoting mitochondrial ROS generation. Chem Biol Interact 2024; 399:111152. [PMID: 39025289 DOI: 10.1016/j.cbi.2024.111152] [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: 04/11/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Cadmium (Cd), a prevalent environmental contaminant, has attracted widespread attention due to its serious health hazards. Ferroptosis is a form of iron-dependent oxidative cell death that contributes to the development of various kidney diseases. However, the mechanisms underlying the occurrence of ferroptosis in Cd-induced renal tubular epithelial cells (TECs) have not been fully elucidated. Hereby, both in-vitro and in-vivo experiments were established to elucidate this issue. In this study, we found that Cd elicited accumulation of lipid peroxides due to intracellular ferrous ion (Fe2+) overload and glutathione depletion, contributing to ferroptosis. Inhibition of ferroptosis via chelation of Fe2+ or reduction of lipid peroxidation can significantly mitigate Cd-induced cytotoxicity. Renal transcriptome analysis revealed that the activation of heme oxygenase 1 (HO-1) was closely related to ferroptosis in Cd-induced TECs injury. Cd-induced ferroptosis and resultant TECs injury are significantly alleviated due to HO-1 inhibition, demonstrating the crucial role of HO-1 in Cd-triggered ferroptosis. Further studies showed that accumulation of lipid peroxides due to iron overload and mitochondrial ROS (mtROS) generation was responsible for HO-1-triggered ferroptosis in Cd-induced cytotoxicity. In conclusion, the current study demonstrates that excessively upregulating HO-1 promotes iron overload and mtROS overproduction to trigger ferroptosis in Cd-induced TECs injury, highlighting that targeting HO-1-mediated ferroptosis may provide new ideas for preventing Cd-induced nephrotoxicity.
Collapse
Affiliation(s)
- Yan-Ting Lv
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 6l Daizong Street, Tai'an City, Shandong Province, 271018, China
| | - Tian-Bin Liu
- New Drug Evaluation Center of Shandong Academy of Pharmaceutical Sciences, Shandong Academy of Pharmaceutical Sciences, 989 Xinluo Street, Ji'nan City 250101 Shandong Province, China
| | - Yue Li
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 6l Daizong Street, Tai'an City, Shandong Province, 271018, China
| | - Zhen-Yong Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 6l Daizong Street, Tai'an City, Shandong Province, 271018, China
| | - Cai-Yu Lian
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 6l Daizong Street, Tai'an City, Shandong Province, 271018, China.
| | - Lin Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 6l Daizong Street, Tai'an City, Shandong Province, 271018, China.
| |
Collapse
|
45
|
Li J, Chen D, Suo J, Li J, Zhang Y, Wang Y, Deng Z, Zhang Q, Ma B. Triptolide induced spermatogenesis dysfunction via ferroptosis activation by promoting K63-linked GPX4 polyubiquitination in spermatocytes. Chem Biol Interact 2024; 399:111130. [PMID: 38960301 DOI: 10.1016/j.cbi.2024.111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024]
Abstract
Triptolide (TP) is a major bioactive compound derived from Tripterygium wilfordii Hook. F. (TwHF) known for its medicinal properties, but it also exhibits potential toxic effects. It has been demonstrated to induce severe male reproductive toxicity, yet the precise mechanism behind this remains unclear, which limits its broad clinical application. This study aimed to investigate the mechanisms underlying testicular damage and spermatogenesis dysfunction induced by TP in mice, using both mouse models and the spermatocyte-derived cell line GC-2spd. In the present study, it was found that TP displayed significant testicular microstructure damaged and spermatogenesis defects including lower concentration and abnormal morphology by promoting ROS formation, MDA production and restraining GSH level, glutathione peroxidase 4 (GPX4) expression in vivo. Furthermore, Ferrostatin-1 (FER-1), a ferroptosis inhibitor, was found to significantly reduce the accumulation of lipid peroxidation, alleviate testicular microstructural damage, and enhance spermatogenic function in mice. Besides, notably decreased cell viability, collapsed mitochondrial membrane potential, and elevated DNA damage were observed in vitro. The above-mentioned phenomenon could be reversed by pre-treatment of FER-1, indicating that ferroptosis participated in the TP-mediated spermatogenesis dysfunction. Mechanistically, TP could enhance GPX4 ubiquitin degradation via triggering K63-linked polyubiquitination of GPX4, thereby stimulating ferroptosis in spermatocytes. Functionally, GPX4 deletion intensified ferroptosis and exacerbated DNA damage in GC-2 cells, while GPX4 overexpression mitigated ferroptosis induced by TP. Overall, these findings for the first time indicated a vital role of ferroptosis in TP induced-testicular injury and spermatogenic dysfunction through promoting GPX4 K63-linked polyubiquitination, which hopefully offers a potential therapeutic avenue for TP-related male reproductive damage. In addition, this study also provides a theoretical foundation for the improved clinical application of TP or TwHF in the future.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Dezhi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Jialiang Suo
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Jiaqi Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Yimu Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Yu Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Zhewen Deng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China.
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
46
|
Zheng S, Jiang J, Shu Z, Qiu C, Jiang L, Zhao N, Lin X, Qian Y, Liang B, Qiu L. Fine particulate matter (PM 2.5) induces testosterone disruption by triggering ferroptosis through SIRT1/HIF-1α signaling pathway in male mice. Free Radic Biol Med 2024; 221:40-51. [PMID: 38759901 DOI: 10.1016/j.freeradbiomed.2024.05.026] [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: 03/20/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Fine particulate matter (PM2.5), a significant component of air pollution particulate matter, is inevitable and closely associated with increasing male reproductive disorder. However, the testicular targets of PM2.5 and its toxicity related molecular mechanisms are still not fully understood. In this study, the conditional knockout (cKO) mice and primary Leydig cells were used to explore the testicular targets of PM2.5 and the related underlying mechanisms. First, apparent the structure impairment of seminiferous tubules, Leydig cells vacuolization, decline of serum testosterone and sperm quality reduction were found in male wild-type (WT) and Sirt1 knockout mice after exposure to PM2.5. Enrichment analyses revealed that differentially expressed genes (DEGs) were enriched in steroid hormone biosynthesis, ferroptosis, and HIF-1 signaling pathway in the mice testes after exposure to PM2.5, which were subsequently verified by the molecular biological analyses. Notably, similar enrichment analyses results were also observed in primary Leydig cells after treatment with PM2.5. In addition, Knockdown of Sirt1 significantly increased PM2.5-induced expression and activation of HIF-1α, which was in parallel to the changes of cellular iron levels, oxidative stress indicators and the ferroptosis markers. In conclusion, this highlights that PM2.5 triggers ferroptosis via SIRT1/HIF-1α signaling pathway to inhibit testosterone synthesis in males. These findings provide a novel research support for the study that PM2.5 causes male reproductive injury.
Collapse
Affiliation(s)
- Shaokai Zheng
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Jinchen Jiang
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Zhenhao Shu
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Chong Qiu
- Medical School, Nantong University, 19 Qixiu Rd, Nantong, 226001, PR China
| | - Lianlian Jiang
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Nannan Zhao
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Xiaojun Lin
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Yingyun Qian
- Graduate School, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Bo Liang
- Department of Ultrasound, Affiliated Hospital 2 of Nantong University, 6 Hai'er Lane North Rd, Nantong, 226019, PR China.
| | - Lianglin Qiu
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China.
| |
Collapse
|
47
|
Li Y, Tian Y, Xu M, Qiu X, Bao Z, Shi M, Deng F, Chen Y, Tang S, Wan Y, Jia X, Yang H. Single-cell insights into mouse testicular toxicity under peripubertal exposure to di(2-ethylhexyl) phthalate. Toxicol Sci 2024; 200:287-298. [PMID: 38730545 DOI: 10.1093/toxsci/kfae064] [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] [Indexed: 05/13/2024] Open
Abstract
Male fertility depends on normal pubertal development. Di-(2-ethylhexyl) phthalate (DEHP) is a potent antiandrogen chemical, and exposure to DEHP during peripuberty can damage the developing male reproductive system, especially the testis. However, the specific cellular targets and differentiation processes affected by DEHP, which lead to testicular toxicity, remain poorly defined. Herein, we presented the first single-cell transcriptomic profile of the pubertal mouse testis following DEHP exposure. To carry out the experiment, 2 groups (n = 8 each) of 3-week-old male mice were orally administered 0.5% carboxymethylcellulose sodium salt or 100 mg/kg body weight DEHP daily from postnatal day 21-48, respectively. Using single-cell RNA sequencing, a total of 31 distinct cell populations were identified, notably, Sertoli and Leydig cells emerged as important targets of DEHP. DEHP exposure significantly decreased the proportions of Sertoli cell clusters expressing mature Sertoli markers (Sox9 and Ar), and selectively reduced the expression of testosterone synthesis genes in fetal Leydig cells. Through cell-cell interaction analyses, we observed changed numbers of interactions in Sertoli cells 1 (SCs1), Leydig cells 1 (LCs1), and interstitial macrophages, and we also identified cell-specific ligand gene expressions in these clusters, such as Inha, Fyn, Vcam1, and Apoe. Complementary in vitro assays confirmed that DEHP directly reduced the expression of genes related to Sertoli cell adhesion and intercellular communication. In conclusion, peripubertal DEHP exposure reduced the number of mature Sertoli cells and may disrupt testicular steroidogenesis by affecting the testosterone synthesis genes in fetal Leydig cells rather than adult Leydig cells.
Collapse
Affiliation(s)
- Yongning Li
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Yaru Tian
- Guangdong Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Food Safety and Health Research Center, Guangzhou 510515, China
| | - Miao Xu
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuemei Qiu
- Reproductive Medicine Center, Zaozhuang Maternal and Child Health Care Hospital, Shandong 277100, China
| | - Zhongjian Bao
- Reproductive Medicine Center, Zaozhuang Maternal and Child Health Care Hospital, Shandong 277100, China
| | - Miaoying Shi
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Fuchang Deng
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Yuanyuan Chen
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Song Tang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yi Wan
- Laboratory for Earth Surface Processes, College of 646 Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Xudong Jia
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Hui Yang
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China
- Guangdong Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Food Safety and Health Research Center, Guangzhou 510515, China
| |
Collapse
|
48
|
Khodasevich D, Holland N, Harley KG, Eskenazi B, Barcellos LF, Cardenas A. Prenatal exposure to environmental phenols and phthalates and altered patterns of DNA methylation in childhood. ENVIRONMENT INTERNATIONAL 2024; 190:108862. [PMID: 38972116 PMCID: PMC11620025 DOI: 10.1016/j.envint.2024.108862] [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: 09/11/2023] [Revised: 06/13/2024] [Accepted: 06/28/2024] [Indexed: 07/09/2024]
Abstract
INTRODUCTION Epigenetic marks are key biomarkers linking the prenatal environment to health and development. However, DNA methylation associations and persistence of marks for prenatal exposure to multiple Endocrine Disrupting Chemicals (EDCs) in human populations have not been examined in great detail. METHODS We measured Bisphenol-A (BPA), triclosan, benzophenone-3 (BP3), methyl-paraben, propyl-paraben, and butyl-paraben, as well as 11 phthalate metabolites, in two pregnancy urine samples, at approximately 13 and 26 weeks of gestation in participants of the Center for the Health Assessment of Mothers and Children of Salinas (CHAMACOS) study (N = 309). DNA methylation of cord blood at birth and child peripheral blood at ages 9 and 14 years was measured with 450K and EPIC arrays. Robust linear regression was used to identify differentially methylated probes (DMPs), and comb-p was used to identify differentially methylated regions (DMRs) in association with pregnancy-averaged EDC concentrations. Quantile g-computation was used to assess associations of the whole phenol/phthalate mixture with DMPs and DMRs. RESULTS Prenatal BPA exposure was associated with 1 CpG among males and Parabens were associated with 10 CpGs among females at Bonferroni-level significance in cord blood. Other suggestive DMPs (unadjusted p-value < 1 × 10-6) and several DMRs associated with the individual phenols and whole mixture were also identified. A total of 10 CpG sites at least suggestively associated with BPA, Triclosan, BP3, Parabens, and the whole mixture in cord blood were found to persist into adolescence in peripheral blood. CONCLUSIONS We found sex-specific associations between prenatal phenol exposure and DNA methylation, particularly with BPA in males and Parabens in females. Additionally, we found several DMPs that maintained significant associations with prenatal EDC exposures at age 9 and age 14 years.
Collapse
Affiliation(s)
- Dennis Khodasevich
- Division of Environmental Health Sciences, Berkeley Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Nina Holland
- Center for Environmental Research and Community Health (CERCH), Berkeley Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Kim G Harley
- Center for Environmental Research and Community Health (CERCH), Berkeley Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Brenda Eskenazi
- Center for Environmental Research and Community Health (CERCH), Berkeley Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Lisa F Barcellos
- Division of Epidemiology, Berkeley Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Andres Cardenas
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
49
|
Mao Z, Ji Q, Chen P, Zhong K, Zeng X. Hydrogen sulfide protects against toxicant acrolein-induced ferroptotic cell death in Sertoli cells. Front Pharmacol 2024; 15:1440147. [PMID: 39148534 PMCID: PMC11324607 DOI: 10.3389/fphar.2024.1440147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
Acrolein (ACR) is a ubiquitous environmental pollutant and byproduct of lipid peroxidation that has been implicated in male infertility. However, the molecular mechanisms underlying ACR-induced toxicity in Sertoli cells remain unclear. Given its role in inducing oxidative stress, we examined whether ferroptosis, an iron-dependent form of regulated cell death, could mediate ACR toxicity in Sertoli cells. We also tested if hydrogen sulfide (H2S), which has antioxidant and ACR detoxifying properties, could protect Sertoli cells from ACR-induced ferroptosis. ACR exposure decreased Sertoli cell viability, increased protein carbonylation and p38 MAPK phosphorylation, indicating oxidative injury. ACR also depleted glutathione (GSH), downregulated the cystine importer SLC7A11, increased intracellular ferrous iron (Fe2+) and lipid peroxidation, suggesting activation of ferroptosis. Consistently, the ferroptosis inhibitor deferoxamine (DFO) markedly attenuates ACR-induced cell death. Further studies revealed that ACR-induced ferroptotic changes were prevented by exogenous H2S and exaggerated by inhibition of endogenous H2S production. Furthermore, H2S also suppressed GPX4 inhibitor RSL3-induced intracellular ACR accumulation and ferroptosis. In summary, our study demonstrates that ACR induces ferroptotic cell death in Sertoli cells, which can be prevented by H2S through multiple mechanisms. Targeting the H2S pathway may represent a therapeutic strategy to mitigate ACR-induced Sertoli cell injury and preserve male fertility.
Collapse
Affiliation(s)
- Zhimin Mao
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu, China
| | - Qun Ji
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu, China
| | - Ping Chen
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu, China
| | - Kun Zhong
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu, China
| | - Xuhui Zeng
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
50
|
Cheng J, Yang L, Zhang Z, Xu D, Hua R, Chen H, Li X, Duan J, Li Q. Diquat causes mouse testis injury through inducing heme oxygenase-1-mediated ferroptosis in spermatogonia. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116562. [PMID: 38850704 DOI: 10.1016/j.ecoenv.2024.116562] [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: 03/08/2024] [Revised: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
Diquat dibromide (DQ) is a globally used herbicide in agriculture, and its overuse poses an important public health issue, including male reproductive toxicity in mammals. However, the effects and molecular mechanisms of DQ on testes are limited. In vivo experiments, mice were intraperitoneally injected with 8 or 10 mg/kg/ day of DQ for 28 days. It has been found that heme oxygenase-1 (HO-1) mediates DQ-induced ferroptosis in mouse spermatogonia, thereby damaging testicular development and spermatogenesis. Histopathologically, we found that DQ exposure caused seminiferous tubule disorders, reduced germ cells, and increased sperm malformation, in mice. Reactive oxygen species (ROS) staining of frozen section and transmission electron microscopy (TEM) displayed DQ promoted ROS generation and mitochondrial morphology alterations in mouse testes, suggesting that DQ treatment induced testicular oxidative stress. Subsequent RNA-sequencing further showed that DQ treatment might trigger ferroptosis pathway, attributed to disturbed glutathione metabolism and iron homeostasis in spermatogonia cells in vitro. Consistently, results of western blotting, measurements of MDA and ferrous iron, and ROS staining confirmed that DQ increased oxidative stress and lipid peroxidation, and accelerated ferrous iron accumulation both in vitro and in vivo. Moreover, inhibition of ferroptosis by deferoxamine (DFO) markedly ameliorated DQ-induced cell death and dysfunction. By RNA-sequencing, we found that the expression of HO-1 was significantly upregulated in DQ-treated spermatogonia, while ZnPP (a specific inhibitor of HO-1) blocked spermatogonia ferroptosis by balancing intracellular iron homeostasis. In mice, administration of the ferroptosis inhibitor ferrostatin-1 effectively restored the increase of HO-1 levels in the spermatogonia, prevented spermatogonia death, and alleviated the spermatogenesis disorders induced by DQ. Overall, these findings suggest that HO-1 mediates DQ-induced spermatogonia ferroptosis in mouse testes, and targeting HO-1 may be an effective protective strategy against male reproductive disorders induced by pesticides in agriculture.
Collapse
Affiliation(s)
- Jianyong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Li Yang
- Health Management Center, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Zelin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Dejun Xu
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Rongmao Hua
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518000, China
| | - Huali Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621000, China
| | - Xiaoya Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Jiaxin Duan
- College of Animal Science, Shanxi Agricultural University, Taiyuan 030801, China
| | - Qingwang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|