1
|
Yan S, Ma H, Ren Y, Wang P, Liu D, Ding N, Liu Y, Chen Q, Ren S, Mou Y. Perfluorooctane sulfonate causes HK-2 cell injury through ferroptosis and endoplasmic reticulum stress pathways. Toxicol Ind Health 2025; 41:73-82. [PMID: 39560653 DOI: 10.1177/07482337241300722] [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: 11/20/2024]
Abstract
Perfluorooctane sulfonate (PFOS) is a synthetic persistent organic compound that is widely used in industrial products. Studies have shown that PFOS can accumulate in environment and pose a threat to human health. As the kidney is the main excretory organ for PFOS, it is important to study PFOS damage to the kidney to investigate its toxicity. Human proximal tubular epithelial cells (HK-2) were treated with 200 μM PFOS or 1 μM Fer-1. Cell viability, the levels of MDA, GSH, intracellular iron ion, and GPX-4 were determined. The expression of KIM-1 and endoplasmic reticulum stress (ERS) related proteins were determined. The expression levels of KIM-1, a marker of renal tubular injury, and ERS-related proteins, GRP78, ATF6, IRE1, and PERK, were significantly increased in HK-2 cells exposed to PFOS. The levels of MDA and intracellular total iron ion also were significantly increased in HK-2 cells exposed to PFOS and the levels of GSH and GPX-4 were significantly decreased. PFOS can damage HK-2 cells through ferroptosis and endoplasmic reticulum stress, which provides a theoretical foundation for exploring the toxicity of PFOS to the kidney.
Collapse
Affiliation(s)
- Shuqi Yan
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Haoyan Ma
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Yuwan Ren
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Pingwei Wang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Dongge Liu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Na Ding
- High-Tech Area Good Doctor Friendship Comprehensive Outpatient Department, Changchun, China
| | - Yanping Liu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Qianqian Chen
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Shuping Ren
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Yan Mou
- Department of Dermatology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Hanvoravongchai J, Laochindawat M, Kimura Y, Mise N, Ichihara S. Clinical, histological, molecular, and toxicokinetic renal outcomes of per-/polyfluoroalkyl substances (PFAS) exposure: Systematic review and meta-analysis. CHEMOSPHERE 2024; 368:143745. [PMID: 39542374 DOI: 10.1016/j.chemosphere.2024.143745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are synthetic chemicals present in the environment that can negatively affect health. Kidney is the major target organ of PFAS exposure, yet the renal impact of PFAS is not completely understood. Here we review the effects of PFAS exposure on kidney health to identify gaps in our understanding and mark potential avenues for future research. METHODS PubMed and SCOPUS databases were searched for studies that examined the association between PFAS exposure and kidney-related outcomes. We included all epidemiological, animal, and cell studies and categorized outcomes into four categories: clinical, histological, molecular and toxicokinetic. RESULTS We identified 169 studies, including 51 on clinical outcomes, 28 on histological changes, 42 on molecular mechanisms, and 68 on toxicokinetics. Perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) exposure were associated with kidney dysfunction, chronic kidney diseases, and increased risk of kidney cancer. Various histological changes were reported, especially in tubular epithelial cells, and the etiology of PFAS-induced kidney injury included various molecular mechanisms. Although PFOA and PFOS are not considered genotoxic, they exhibit several characteristics of carcinogens. Toxicokinetics of PFOA and PFOS differed significantly between species, with renal elimination influenced by various factors such as sex, age, and structure of the compound. CONCLUSION Evidence suggests that PFAS, especially PFOA and PFOS, negatively affects kidney health, though gaps in our understanding of such effects call for further research.
Collapse
Affiliation(s)
- Jidapa Hanvoravongchai
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Methasit Laochindawat
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yusuke Kimura
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Nathan Mise
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan.
| |
Collapse
|
3
|
Li Y, Lv Y, Jiang Z, Ma C, Li R, Zhao M, Guo Y, Guo H, Zhang X, Li A, Liu Y. Association of co-exposure to organophosphate esters and per- and polyfluoroalkyl substances and mixture with cardiovascular-kidney-liver-metabolic biomarkers among Chinese adults. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116524. [PMID: 38838464 DOI: 10.1016/j.ecoenv.2024.116524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Organophosphate esters (OPEs) and Per- and polyfluoroalkyl substances (PFAS) are ubiquitous environmental contaminants with common exposure sources, leading to their widespread presence in human body. However, evidence on co-exposure to OPEs and PFAS and its impact on cardiovascular-kidney-liver-metabolic biomarkers remains limited. METHODS In this cross-sectional study, 467 adults were enrolled from January to May 2022 during physical visits in Shijiazhuang, Hebei province. Eleven types of OPEs and twelves types of PFAS were detected, among which eight OPEs and six PFAS contaminants were detected in more than 60% of plasma samples. Seventeen biomarkers were assessed to comprehensively evaluate the cardiovascular-kidney-liver-metabolic function. Multiple linear regression, multipollutant models with sparse partial least squares, and Bayesian kernel machine regression (BKMR) models were applied to examine the associations of individual OPEs and PFAS and their mixtures with organ function and metabolism, respectively. RESULTS Of the over 400 exposure-outcome associations tested when modelling, we observed robust results across three models that perfluorohexanoic acid (PFHxS) was significantly positively associated with alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), and indirect bilirubin (IBIL). Perfluorononanoic acid was significantly associated with decreased AST/ALT and increased very-low-density lipoprotein cholesterol levels. Besides, perfluorodecanoic acid was correlated with increased high lipoprotein cholesterol and perfluoroundecanoic acid was consistently associated with lower glucose level. BKMR analysis showed that OPEs and PFAS mixtures were positively associated with IBIL and TBIL, among which PFHxS was the main toxic chemicals. CONCLUSIONS Our findings suggest that exposure to OPEs and PFAS, especially PFHxS and PFNA, may disrupt organ function and metabolism in the general population, providing insight into the potential pathophysiological mechanisms of OPEs and PFAS co-exposure and chronic diseases.
Collapse
Affiliation(s)
- Yanbing Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, PR China
| | - Yi Lv
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Zexuan Jiang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Chaoying Ma
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Ran Li
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Mengwei Zhao
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yi Guo
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Huicai Guo
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei Province 050017, PR China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, PR China
| | - Xiaoguang Zhang
- Core Facilities and Centers of Hebei Medical University, Shijiazhuang 050017, PR China
| | - Ang Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei Province 050017, PR China.
| | - Yi Liu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei Province 050017, PR China.
| |
Collapse
|
4
|
Li X, Ma TK, Wang P, Shi H, Hai S, Qin Y, Zou Y, Zhu WT, Li HM, Li YN, Yin L, Xu YY, Yang Q, Zhang S, Ding H. HOXD10 attenuates renal fibrosis by inhibiting NOX4-induced ferroptosis. Cell Death Dis 2024; 15:398. [PMID: 38844470 PMCID: PMC11156659 DOI: 10.1038/s41419-024-06780-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024]
Abstract
In chronic kidney disease (CKD), renal fibrosis is an unavoidable result of various manifestations. However, its pathogenesis is not yet fully understood. Here, we revealed the novel role of Homeobox D10 (HOXD10) in CKD-related fibrosis. HOXD10 expression was downregulated in CKD-related in vitro and in vivo fibrosis models. UUO model mice were administered adeno-associated virus (AAV) containing HOXD10, and HOXD10 overexpression plasmids were introduced into human proximal tubular epithelial cells induced by TGF-β1. The levels of iron, reactive oxygen species (ROS), lipid ROS, the oxidized glutathione/total glutathione (GSSG/GSH) ratio, malonaldehyde (MDA), and superoxide dismutase (SOD) were determined using respective assay kits. Treatment with AAV-HOXD10 significantly attenuated fibrosis and renal dysfunction in UUO model mice by inhibiting NOX4 transcription, ferroptosis pathway activation, and oxidative stress. High levels of NOX4 transcription, ferroptosis pathway activation and profibrotic gene expression induced by TGF-β1/erastin (a ferroptosis agonist) were abrogated by HOXD10 overexpression in HK-2 cells. Moreover, bisulfite sequencing PCR result determined that HOXD10 showed a hypermethylated level in TGF-β1-treated HK-2 cells. The binding of HOXD10 to the NOX4 promoter was confirmed by chromatin immunoprecipitation (ChIP) analysis and dual-luciferase reporter assays. Targeting HOXD10 may represent an innovative therapeutic strategy for fibrosis treatment in CKD.
Collapse
Affiliation(s)
- Xin Li
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Tian-Kui Ma
- Biological Therapy Department, First Hospital of China Medical University, Shenyang, China
| | - Pu Wang
- General Practice Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Hang Shi
- Intensive Care Unit Department, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Sang Hai
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Yu Qin
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Yun Zou
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Wan-Ting Zhu
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Hui-Min Li
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Yan-Nong Li
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Li Yin
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Yan-Yan Xu
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Qi Yang
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Shuang Zhang
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China
| | - Hong Ding
- Nephrology Department, Fourth Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
5
|
Mokra K, Kaczmarska I, Bukowska B. Perfluorooctane sulfonate (PFOS) and its selected analogs induce various cell death types in peripheral blood mononuclear cells. CHEMOSPHERE 2024; 354:141664. [PMID: 38485001 DOI: 10.1016/j.chemosphere.2024.141664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024]
Abstract
The perfluoalkyl substance (PFASs) perfluorooctane sulfonate (PFOS) has been widely used in industry. However, PFOS is a persistent organic pollutant and has been gradually replaced by its short-chain analogs, perfluorohexane sulfonate (PFHxS) and perfluorobutane sulfonate (PFBS). PFASs are extremely persistent and are very frequently detected among the general population. The aim of the study was to determine the effect of selected PFASs on peripheral blood mononuclear cells (PBMCs) and the mechanisms of their action. PBMCs were exposed to PFOS, PFBS and PFHxS at concentrations ranging from 0.02 to 400 μM for 24 h, they were then tested for viability, apoptosis (changes in cytosolic calcium ions level and caspase-3, -8 and -9 activation), ferroptosis (changes in chelatable iron ions level and lipid peroxidation), and autophagy (LC3-II and Raptor level assay). PFOS exposure decreased cell viability, increased calcium ion level and caspase-8 activation; it also enhanced lipid peroxidation and increased the intracellular pool of chelatable iron ions as well as LC3-II protein content. In contrast, short-chain PFBS and PFHxS induced significant changes in the markers of apoptosis but had no substantial impact on ferroptosis or autophagy markers over a wide range of concentrations. Our results indicate that only PFOS demonstrated pro-ferroptotic and pro-autophagic potential but observed changes occurred at relatively high exposure. A short-chain substitute (PFBS) exhibited strong pro-apoptotic potential at concentrations related to occupational exposure. While the short-chain PFASs strongly affected the mitochondrial pathway of apoptosis, apoptosis itself was only induced by PFBS via the intrinsic and extrinsic pathways. It seems that the length of the carbon chain in PFASs appears to determine the cell death mechanisms activated in human PBMCs following exposure. Our findings provide a new insight into the immune toxicity mechanism induced by these compounds.
Collapse
Affiliation(s)
- Katarzyna Mokra
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Biophysics of Environmental Pollution, 141/143 Pomorska St., 90-236, Lodz, Poland.
| | - Izabela Kaczmarska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Biophysics of Environmental Pollution, 141/143 Pomorska St., 90-236, Lodz, Poland
| | - Bożena Bukowska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Biophysics of Environmental Pollution, 141/143 Pomorska St., 90-236, Lodz, Poland
| |
Collapse
|
6
|
Zhu H, Duan Y, Yang Y, Chen E, Huang H, Wang X, Zhou J. Sodium aescinate induces renal toxicity by promoting Nrf2/GPX4-mediated ferroptosis. Chem Biol Interact 2024; 391:110892. [PMID: 38364601 DOI: 10.1016/j.cbi.2024.110892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 02/18/2024]
Abstract
Sodium aescinate (SA) is extracted from Aesculus wilsonii Rehd seeds and was first marketed as a medicament in German. With the wide application of SA in clinical practice, reports of adverse drug reactions and adverse events have gradually increased, including renal impairment. However, the pathogenic mechanisms of SA have not yet been fully elucidated. The toxic effects and underlying mechanisms of SA were explored in this study. Our data showed that SA significantly elevated the levels of blood urea nitrogen (BUN), serum creatinine (Scr) and Kidney injury molecule 1 (Kim-1), accompanied by pathologically significant changes in renal tissue. SA induced NRK-52E cell death and disrupted the integrity of the cell membrane. Moreover, SA caused significant reductions in FTH, Nrf2, xCT, GPX4, and FSP1 levels, but increased TFR1 and ACSL4 levels. SA decreased glutathione peroxidase (GPx), glutathione (GSH) and cysteine (Cys) levels, but improved Fe2+, malondialdehyde (MDA), reactive oxygen species (ROS) and lipid peroxidation levels, ultimately leading to the induction of ferroptosis. Importantly, inhibition of ferroptosis or activation of the Nrf2/GPX4 pathway prevented SA-induced nephrotoxicity. These findings indicated that SA induced oxidative damage and ferroptosis-mediated kidney injury by suppressing the Nrf2/GPX4 axis activity.
Collapse
Affiliation(s)
- Haiyan Zhu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Yenan Duan
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Yijing Yang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Enqing Chen
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Hanxin Huang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Xi Wang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China.
| |
Collapse
|
7
|
Yang L, Cai X, Li R. Ferroptosis Induced by Pollutants: An Emerging Mechanism in Environmental Toxicology. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:2166-2184. [PMID: 38275135 DOI: 10.1021/acs.est.3c06127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Environmental pollutants have been recognized for their ability to induce various adverse outcomes in both the environment and human health, including inflammation, apoptosis, necrosis, pyroptosis, and autophagy. Understanding these biological mechanisms has played a crucial role in risk assessment and management efforts. However, the recent identification of ferroptosis as a form of programmed cell death has emerged as a critical mechanism underlying pollutant-induced toxicity. Numerous studies have demonstrated that fine particulates, heavy metals, and organic substances can trigger ferroptosis, which is closely intertwined with lipid, iron, and amino acid metabolism. Given the growing evidence linking ferroptosis to severe diseases such as heart failure, chronic obstructive pulmonary disease, liver injury, Parkinson's disease, Alzheimer's disease, and cancer, it is imperative to investigate the role of pollutant-induced ferroptosis. In this review, we comprehensively analyze various pollutant-induced ferroptosis pathways and intricate signaling molecules and elucidate their integration into the driving and braking axes. Furthermore, we discuss the potential hazards associated with pollutant-induced ferroptosis in various organs and four representative animal models. Finally, we provide an outlook on future research directions and strategies aimed at preventing pollutant-induced ferroptosis. By enhancing our understanding of this novel form of cell death and developing effective preventive measures, we can mitigate the adverse effects of environmental pollutants and safeguard human and environmental health.
Collapse
Affiliation(s)
- Lili Yang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xiaoming Cai
- School of Public Health, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ruibin Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
8
|
Li X, Zhu S, Li Z, Meng Y, Huang S, Yu Q, Li B. Melittin induces ferroptosis and ER stress-CHOP-mediated apoptosis in A549 cells. Free Radic Res 2022; 56:398-410. [PMID: 36194238 DOI: 10.1080/10715762.2022.2131551] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Melittin is a natural polypeptide present in bee venom, with significant anti-tumor activity. Melittin has been reported to induce cell death in lung carcinoma cell line A549 cells, suggesting an excellent potential for treating lung cancer. However, the core mechanism underlying melittin-induced cell death in A549 cells remains unclear. This work reports that melittin induces reactive oxygen species (ROS) burst, upregulates intracellular Fe2+ levels, disrupts the glutathione-glutathione peroxidase 4 antioxidant system, and increases lipid peroxide accumulation, eventually inducing cell death, indicating that ferroptosis may be involved in the antitumor effects of melittin in A549 cells. Furthermore, A549 cells treated with the ferroptosis inhibitors ferrostatin-1 and deferoxamine demonstrated that these inhibitors could reverse the cell death induced by melittin, further confirming that melittin induces A549 cell death via ferroptosis. Furthermore, the results also illustrated that melittin activated the endoplasmic reticulum (ER) stress-CHOP (C/EBP homologous protein) apoptotic signal, closely associated with high-level intracellular ROS. The ER stress inhibitor, 4-Phenyl butyric acid, was used to confirm that ER stress-CHOP apoptotic signaling is another molecular mechanism of melittin-induced A549 cell death. Thus, our results demonstrate that ferroptosis and ER stress-CHOP signaling are key molecular mechanisms of melittin-induced cell death in lung cancer.Key policy highlightsMelittin upregulates intracellular Fe2+ levels, leading to the accumulation of lipid peroxides in A549 cells.Melittin disrupts the glutathione-glutathione peroxidase 4 antioxidant system in A549 cells.Melittin induces activation of endoplasmic reticulum stress-C/EBP homologous protein apoptosis signal.Ferroptosis and ER stress are the core molecular mechanisms underlying melittin-induced apoptosis in A549 cells.
Collapse
Affiliation(s)
- Xuan Li
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| | - Sen Zhu
- School of Life Sciences, Lanzhou University, 730030, Lanzhou, China
| | - Zheng Li
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| | - Yuqi Meng
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| | - Sujie Huang
- School of Basic Medical Sciences, Lanzhou University, 730030, Lanzhou, China
| | - Qiyao Yu
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| | - Bin Li
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| |
Collapse
|