1
|
Liu N, Liu G, Li Q, Hu Y, Wang H. Effects on Iron Metabolism and System Xc- /GPX4 Pathway from Hydroquinone Suggest Ferroptosis of Jurkat Cells. TOXICS 2024; 12:644. [PMID: 39330572 PMCID: PMC11436165 DOI: 10.3390/toxics12090644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/28/2024]
Abstract
Prolonged exposure to hydroquinone (HQ), a metabolite of benzene, can cause severe haematologic disorders in humans. However, the mechanism is still unclear. In the present study, we investigated whether HQ can induce haematological diseases through ferroptosis, which is another form of cell death apart from apoptosis. The results showed that HQ inhibited the viability of Jurkat cells in a dose-dependent and time-dependent manner. The half inhibitory concentrations (IC50s) of HQ-treated Jurkat cells for 12 h, 24 h and 48 h were 107.16 μmol/L, 33.29 μmol/L, and 14.78 μmol/L. The exposure of Jurkat cells to HQ increased intracellular Fe2+, malondialdehyde (MDA) and lipid reactive oxygen species (ROS) levels and down-regulated glutathione (GSH) levels. We used erastin-treated cells as a positive control and cells treated with HQ combined with deferoxamine mesylate (DFO) and ferrostain-1 (Fer-1)-treated cells as the negative controls. DFO and Fer-1 partially restored the degradation of cell viability and GSH content and the accumulation of Fe2+, MDA and lipid ROS caused by HQ. In addition, we found that cellular mitochondria in the HQ-treated group showed a decrease in volume, an increase in the density of the bilayer membrane and a decrease or disappearance of mitochondrial cristae. Changes in the erastin-treated group were similar to those in the HQ-treated group. We inferred that HQ induces ferroptosis in Jurkat cells. Subsequently, we found that HQ up-regulated the levels of transferrin receptor 1 (TFRC) mRNA and protein expression and down-regulated FTH1, SLC7A11 and synthetic substrate of antioxidant enzyme 4 (GPX4) mRNA levels and protein expression levels. However, the exposure of Jurkat cells to HQ with DFO and Fer-1 alleviated these changes. Notably, the activation of TFRC and the inhibition of FTH1 and System Xc- (cystine-glutamate reverse transporter protein) /GPX4 were associated with HQ-induced ferroptosis. These results provide novel insights into how HQ exacerbates haematopoietic cytotoxicity and provide potential targets for the prevention of HQ-induced diseases.
Collapse
Affiliation(s)
- Nana Liu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China (Q.L.); (Y.H.)
| | - Ge Liu
- Wuhan Center for Disease Control & Prevention, Wuhan 430000, China;
| | - Qiang Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China (Q.L.); (Y.H.)
| | - Yipeng Hu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China (Q.L.); (Y.H.)
| | - Hong Wang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China (Q.L.); (Y.H.)
| |
Collapse
|
2
|
Xiang Q, Li N, Zhang Y, Wang T, Wang Y, Bian J. GPR116 alleviates acetaminophen-induced liver injury in mice by inhibiting endoplasmic reticulum stress. Cell Mol Life Sci 2024; 81:299. [PMID: 39001944 PMCID: PMC11335223 DOI: 10.1007/s00018-024-05313-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/30/2024] [Accepted: 06/10/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Acetaminophen (APAP) overdose is a significant contributor to drug-induced liver injury worldwide. G-protein-coupled receptor 116 (GPR116) is an important homeostatic maintenance molecule in the body, but little is known about its role in APAP-induced liver injury (AILI). METHODS GPR116 expression was determined in both human and mouse AILI models. Hepatic function and damage response were analyzed in hepatocyte-specific GPR116 deletion (GPR116△HC) mice undergoing APAP challenge. RNA-sequencing, immunofluorescence confocal, and co-immunoprecipitation (CO-IP) were employed to elucidate the impact and underlying mechanisms of GPR116 in AILI. RESULTS Intrahepatic GPR116 was upregulated in human and mice with AILI. GPR116△HC mice were vulnerable to AILI compared to wild-type mice. Overexpression of GPR116 effectively mitigated AILI in wild-type mice and counteracted the heightened susceptibility of GPR116△HC mice to APAP. Mechanistically, GPR116 inhibits the binding immunoglobulin protein (BiP), a critical regulator of ER function, through its interaction with β-arrestin1, thereby mitigating ER stress during the early stage of AILI. Additionally, the activation of GPR116 by ligand FNDC4 has been shown to confer a protective effect against early hepatotoxicity caused by APAP in murine model. CONCLUSIONS Upregulation of GPR116 on hepatocytes inhibits ER stress by binding to β-arrestin1, protecting mice from APAP-induced hepatotoxicity. GPR116 may serve as a promising therapeutic target for AILI.
Collapse
Affiliation(s)
- Qian Xiang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
- Department of Anesthesiology, Peking University Third Hospital, Beijing, 100191, China
| | - Na Li
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yan Zhang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Ting Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Ying Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
3
|
Wu Y, Liu H, Sun Z, Liu J, Li K, Fan R, Dai F, Tang H, Hou Q, Li J, Tang X. The adhesion-GPCR ADGRF5 fuels breast cancer progression by suppressing the MMP8-mediated antitumorigenic effects. Cell Death Dis 2024; 15:455. [PMID: 38937435 PMCID: PMC11211477 DOI: 10.1038/s41419-024-06855-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
ADGRF5 (GPR116) has been identified as a facilitator of breast cancer cell migration and metastasis, yet the underlying mechanisms remain largely elusive. Our current study reveals that the absence of ADGRF5 in breast cancer cells impairs extracellular matrix (ECM)-associated cell motility and impedes in vivo tumor growth. This correlates with heightened expression of matrix metalloproteinase 8 (MMP8), a well-characterized antitumorigenic MMP, and a shift in the polarization of tumor-associated neutrophils (TANs) towards the antitumor N1 phenotype in the tumor microenvironment (TME). Mechanistically, ADGRF5 inhibits ERK1/2 activity by enhancing RhoA activation, leading to decreased phosphorylation of C/EBPβ at Thr235, hindering its nuclear translocation and subsequent activation. Crucially, two C/EBPβ binding motifs essential for MMP8 transcription are identified within its promoter region. Consequently, ADGRF5 silencing fosters MMP8 expression and CXCL8 secretion, attracting increased infiltration of TANs; simultaneously, MMP8 plays a role in decorin cleavage, which leads to trapped-inactivation of TGF-β in the TME, thereby polarizing TANs towards the antitumor N1 neutrophil phenotype and mitigating TGF-β-enhanced cell motility in breast cancer. Our findings reveal a novel connection between ADGRF5, an adhesion G protein-coupled receptor, and the orchestration of the TME, which dictates malignancy progression. Overall, the data underscore ADGRF5 as a promising therapeutic target for breast cancer intervention.
Collapse
Grants
- 82372645 National Natural Science Foundation of China (National Science Foundation of China)
- 81972602 National Natural Science Foundation of China (National Science Foundation of China)
- 82002716 National Natural Science Foundation of China (National Science Foundation of China)
- 82273497 National Natural Science Foundation of China (National Science Foundation of China)
- 81502331 National Natural Science Foundation of China (National Science Foundation of China)
- The Natural Science Foundation of Hunan Province (grant nos. 2023JJ20021), the Fundamental Research Funds for the Central Universities (521119200099, 541109030051).
- The Natural Science Foundation of Hunan Province (grant nos.2024JJ6490)
- Natural Science Foundation of Henan Province (222300420029), Program for Science and Technology Innovation Talents in Universities of Henan Province (23HASTIT042).
- The Project of Department of Education of Guangdong Province, (2019KTSCX146), the Shenzhen Science and Technology Program (JCYJ20190808164209301), the Shenzhen Scientific Research Foundation for Excellent Returned Scholars (000493), the Natural Science Foundation of Shenzhen University General Hospital (SUGH2020QD005), the Disciple gathering teaching project of Shenzhen University, the Shenzhen Key Laboratory Foundation (ZDSYS20200811143757022), the Teaching Reform Research Project of Shenzhen University (YXBJG202339), and the Shenzhen International Cooperation Research Project (GJHZ20220913143004008).
- The Wisdom Accumulation and Talent Cultivation Project of the Third Xiangya Hospital of Central South University (YX202105), Natural Science Foundation of Hunan Province (Grant Nos. 2021JJ31010).
Collapse
Affiliation(s)
- Yalan Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Huixia Liu
- Hunan Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Zhe Sun
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jieling Liu
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Kai Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ronghui Fan
- Hunan Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Fujun Dai
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, 475004, Henan, China
| | - Hui Tang
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, 637003, Sichuan, China
| | - Qi Hou
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of Shenzhen University, Shenzhen, 518061, China
| | - JinSong Li
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Xiaolong Tang
- Hunan Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha, 410082, China.
| |
Collapse
|
4
|
Zhao X, Leng D, Wang H, Jin H, Wu Y, Qin Z, Wu D, Wei X. An Acid-Responsive Iron-Based Nanocomposite for OSCC Treatment. J Dent Res 2024; 103:612-621. [PMID: 38684484 DOI: 10.1177/00220345241238154] [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: 05/02/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of oral cancer, characterized by invasiveness, local lymph node metastasis, and poor prognosis. Traditional treatment and medications have limitations, making the specific inhibition of OSCC growth, invasion, and metastasis a challenge. The tumor microenvironment exhibits mildly acidity and high concentrations of H2O2, and its exploitation for cancer treatment has been widely researched across various cancers, but research in the oral cancer field is relatively limited. In this study, by loading ultra-small Prussian blue nanoparticles (USPBNPs) into mesoporous calcium-silicate nanoparticles (MCSNs), we developed an acid-responsive iron-based nanocomposite, USPBNPs@MCSNs (UPM), for the OSCC treatment. UPM demonstrated excellent dual enzyme activities, generating toxic ·OH in a mildly acidic environment, effectively killing OSCC cells and producing O2 in a neutral environment to alleviate tissue hypoxia. The results showed that UPM could effectively inhibit the proliferation, migration, and invasion of OSCC cells, as well as the growth of mice solid tumors, without obvious systemic toxicity. The mechanisms may involve UPM inducing ferroptosis of OSCC cells by downregulating the xCT/GPX4/glutathione (GSH) axis, characterized by intracellular iron accumulation, reactive oxygen species accumulation, GSH depletion, lipid peroxidation, and abnormal changes in mitochondrial morphology. Therefore, this study provides empirical support for ferroptosis as an emerging therapeutic target for OSCC and offers a valuable insight for future OSCC treatment.
Collapse
Affiliation(s)
- X Zhao
- Jiangsu Province Key Laboratory of Oral Diseases & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine & Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - D Leng
- Jiangsu Province Key Laboratory of Oral Diseases & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine & Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - H Wang
- Jiangsu Province Key Laboratory of Oral Diseases & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine & Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - H Jin
- Jiangsu Province Key Laboratory of Oral Diseases & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine & Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Y Wu
- Jiangsu Province Key Laboratory of Oral Diseases & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine & Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Z Qin
- Nanjing Medical University, The First Clinical Medical College, Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - D Wu
- Jiangsu Province Key Laboratory of Oral Diseases & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine & Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - X Wei
- Jiangsu Province Key Laboratory of Oral Diseases & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine & Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Zhang W, Liu Y, Liao Y, Zhu C, Zou Z. GPX4, ferroptosis, and diseases. Biomed Pharmacother 2024; 174:116512. [PMID: 38574617 DOI: 10.1016/j.biopha.2024.116512] [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: 12/07/2023] [Revised: 03/03/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
GPX4 (Glutathione peroxidase 4) serves as a crucial intracellular regulatory factor, participating in various physiological processes and playing a significant role in maintaining the redox homeostasis within the body. Ferroptosis, a form of iron-dependent non-apoptotic cell death, has gained considerable attention in recent years due to its involvement in multiple pathological processes. GPX4 is closely associated with ferroptosis and functions as the primary inhibitor of this process. Together, GPX4 and ferroptosis contribute to the pathophysiology of several diseases, including sepsis, nervous system diseases, ischemia reperfusion injury, cardiovascular diseases, and cancer. This review comprehensively explores the regulatory roles and impacts of GPX4 and ferroptosis in the development and progression of these diseases, with the aim of providing insights for identifying potential therapeutic strategies in the future.
Collapse
Affiliation(s)
- Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yang Liu
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yan Liao
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Chenglong Zhu
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| | - Zui Zou
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
6
|
Nie A, Shen C, Zhou Z, Wang J, Sun B, Zhu C. Ferroptosis: Potential opportunities for natural products in cancer therapy. Phytother Res 2024; 38:1173-1190. [PMID: 38116870 DOI: 10.1002/ptr.8088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/21/2023]
Abstract
Cancer cells often exhibit defects in the execution of cell death, resulting in poor clinical outcomes for patients with many cancer types. Ferroptosis is a newly discovered form of programmed cell death characterized by intracellular iron overload and lipid peroxidation in the cell membrane. Increasing evidence suggests that ferroptosis is closely associated with a wide variety of physiological and pathological processes, particularly in cancer. Notably, various bioactive natural products have been shown to induce the initiation and execution of ferroptosis in cancer cells, thereby exerting anticancer effects. In this review, we summarize the core regulatory mechanisms of ferroptosis and the multifaceted roles of ferroptosis in cancer. Importantly, we focus on natural products that regulate ferroptosis in cancer cells, such as terpenoids, polyphenols, alkaloids, steroids, quinones, and polysaccharides. The clinical efficacy, adverse effects, and drug-drug interactions of these natural products need to be evaluated in further high-quality studies to accelerate their application in cancer treatment.
Collapse
Affiliation(s)
- Anzheng Nie
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaozan Shen
- Department of Clinical Pharmacy, The Second People's Hospital of Huaihua, Huaihua, China
| | - Zheng Zhou
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Juan Wang
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bao Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chunsheng Zhu
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|