1
|
Zheng J, Liu Y, Zhu F, Liu S, Cai Z, Liu M, An X, Yao Y, Chen N, Guo D. Picropodophyllin induces ferroptosis via blockage of AKT/NRF2/SLC7A11 and AKT/NRF2/SLC40A1 axes in hepatocellular carcinoma as a natural IGF1R inhibitor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156840. [PMID: 40412057 DOI: 10.1016/j.phymed.2025.156840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/15/2025] [Accepted: 05/10/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Ferroptosis represents a distinct form of regulated cell death characterized by intracellular iron overload and extensive lipid peroxidation. Targeting ferroptosis-related signaling pathways and inducing ferroptosis have emerged as promising therapeutic strategies for hepatocellular carcinoma (HCC). Recent studies have highlighted the involvement of insulin-like growth factor 1 receptor (IGF1R) signaling in cancer progression and antioxidant defense mechanisms. Picropodophyllin (PPP), a natural IGF1R inhibitor isolated from Dysosma versipellis, exhibits anticancer effects against several solid tumors. However, the impact of PPP on ferroptosis in HCC and the underlying molecular mechanisms remain unclear. PURPOSE The current study aims to evaluate the anti-tumor effects of PPP on HCC progression in vitro and in vivo, and to investigate the actions and mechanisms of PPP as a novel ferroptosis inducer. METHODS Clinical sample from HCC patients were applied to analyze the correlation of IGF1R with malignancy of HCC. Docking simulations, molecular dynamics simulation and cellular thermal shift assay were performed to verify the interaction between PPP and IGF1R. CCK-8 cell viability assay, colony formation, Calcein-AM/PI staining, wound healing and transwell assays were conducted to determine the effects of PPP on cell viability, proliferation, migration and invasion. Intracellular Fe2+, GSH, MDA and lipid ROS levels were measured to evaluate the degree of ferroptosis induced by PPP. GO functional annotation and KEGG enrichment analysis, quantitative real-time PCR, western blot and immunofluorescence (IF) assay were performed to investigate the mechanisms underlying the action of PPP. Nude mice xenograft model and immunohistochemistry (IHC) assay were utilized to observe the impact of PPP on tumor growth in vivo. RESULTS Upregulation of IGF1R were confirmed to positively correlated with malignant progression of HCC and PPP were verified to act as a specific inhibitor of IGF1R in HCC. PPP exhibited dose-dependent anti-proliferative and anti-metastasis effects on HCC cells, and inhibited HCC growth in a subcutaneous xenograft murine model. Meanwhile, PPP remarkably increased intracellular Fe2+, lipid ROS and MDA levels, but decreased ROS scavenger GSH content and glutathione peroxidase 4 (GPX4) activity significantly, which suggested that PPP stimulated ferroptosis relying on iron-dependent lipid peroxidation. The ferroptosis inhibitor deferoxamine mesylate (DFO) nearly abolished the anti-cancer and ferroptosis-inducing effects of PPP both in vitro and in vivo. Mechanistically, PPP inhibited the phosphorylation of IGF1R, PI3K and AKT, thus suppressed the protein stability of NRF2 by facilitating ubiquitination, and consequently decreased expression of its target gene SLC7A11 and SLC40A1. CONCLUSION The natural IGF1R inhibitor PPP induced ferroptosis through blockage of PI3K/AKT/NRF2 signaling pathway and subsequent inhibition of downstream gene expression of SLC7A11 and SLC40A1 in hepatocellular carcinoma. Consequently, our findings provide a novel action and mechanism of PPP, as well as offer innovative and promising ferroptosis-inducing agents for the clinical treatment of HCC.
Collapse
Affiliation(s)
- Jiahui Zheng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Boulevard (North), Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Guangzhou 510515, China
| | - Yixin Liu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Boulevard (North), Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Guangzhou 510515, China
| | - Fengchi Zhu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Boulevard (North), Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Guangzhou 510515, China
| | - Sha Liu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Boulevard (North), Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Guangzhou 510515, China
| | - Zhuo Cai
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou 510600, China
| | - Mengting Liu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Boulevard (North), Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Guangzhou 510515, China
| | - Xiangping An
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Boulevard (North), Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Guangzhou 510515, China
| | - Yan Yao
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Nana Chen
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou, 510515, China.
| | - Dan Guo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Boulevard (North), Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Guangzhou 510515, China.
| |
Collapse
|
2
|
Yuan X, Yu S, Lin L, Chen Y, Wu Z, Fang X, Zhang W. Brusatol inhibits malignant phenotypes and lipid metabolism of osteosarcoma cells by regulating PI3K/AKT and MAPK pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156464. [PMID: 39970856 DOI: 10.1016/j.phymed.2025.156464] [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: 10/19/2024] [Revised: 01/06/2025] [Accepted: 02/02/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Osteosarcoma (OS), the most frequent type of primary bone cancer, has a poor prognosis in metastatic cases, with overall 5-year survival rates stagnating at 20 %-30 %. This highlights the critical need for innovative therapies to address the significant survival gap between metastatic and non-metastatic cases. Brusatol (BRU), a compound extracted from Brucea javanica, has shown promising anti-tumor properties in various cancers; however, its effects on OS have yet to be investigated. PURPOSE To investigate the anti-tumor mechanisms of BRU in OS and evaluate its potential therapeutic efficacy, with a particular focus on its impact on lipid metabolism and related signaling pathways. METHODS In vitro experiments to assess the anti-tumor effects of BRU involved colony formation, CCK-8, Transwell analysis, as well as flow cytometry. RNA sequencing was conducted to identify transcriptional changes in BRU-treated cells. The mechanism of action was investigated through analysis of lipid metabolism and key signaling pathways. Therapeutic efficacy and safety were evaluated in vivo using xenograft models. RESULTS BRU significantly inhibited OS cell proliferation, migration, and invasion, while also inducing G2/M phase cell cycle arrest as well as promoting apoptosis. Transcriptome analysis revealed that BRU affected lipid metabolism-related genes and suppressed the PI3K/AKT and MAPK pathways. BRU treatment reduced fatty acid synthase expression and free fatty acid content in OS cells. In vivo experiments demonstrated that BRU effectively restricted xenograft growth. CONCLUSION This study revealed that BRU exhibits potent anti-tumor effects in OS by modulating lipid metabolism through the PI3K/AKT and MAPK pathways.
Collapse
Affiliation(s)
- Xuhui Yuan
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Shaolin Yu
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Lan Lin
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Yang Chen
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Zhaoyang Wu
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China
| | - Xinyu Fang
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China.
| | - Wenming Zhang
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China; Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, PR China; Fujian Provincial Institute of Orthopaedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, PR China.
| |
Collapse
|
3
|
Xiong X, Li W, Yu C, Qiu M, Zhang Z, Hu C, Zhu S, Yang L, Pen H, Song X, Chen J, Xia B, Han S, Yang C. SMURF1-Induced Ubiquitination of FTH1 Disrupts Iron Homeostasis and Suppresses Myogenesis. Int J Mol Sci 2025; 26:1390. [PMID: 39941157 PMCID: PMC11818545 DOI: 10.3390/ijms26031390] [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: 12/09/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Ferritin heavy chain 1 (FTH1) is pivotal in the storage, release, and utilization of iron, plays a crucial role in the ferroptosis pathway, and exerts significant impacts on various diseases. Iron influences skeletal muscle development and health by promoting cell growth, ensuring energy metabolism and ATP synthesis, maintaining oxygen supply, and facilitating protein synthesis. However, the precise molecular mechanisms underlying iron's regulation of skeletal muscle growth and development remain elusive. In this study, we demonstrated that the conditional knockout (cKO) of FTH1 in skeletal muscle results in muscle atrophy and impaired exercise endurance. In vitro studies using FTH1 cKO myoblasts revealed notable decreases in GSH concentrations, elevated levels of lipid peroxidation, and the substantial accumulation of Fe2+, collectively implying the induction of ferroptosis. Mechanistically, E3 ubiquitin-protein ligase SMURF1 (SMURF1) acts as an E3 ubiquitin ligase for FTH1, thereby facilitating the ubiquitination and subsequent degradation of FTH1. Consequently, this activation of the ferroptosis pathway by SMURF1 impedes myoblast differentiation into myotubes. This study identifies FTH1 as a novel regulator of muscle cell differentiation and skeletal muscle development, implicating its potential significance in maintaining skeletal muscle health through the regulation of iron homeostasis.
Collapse
Affiliation(s)
- Xia Xiong
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Wen Li
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Chunlin Yu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Mohan Qiu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Zengrong Zhang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Chenming Hu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Shiliang Zhu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Li Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Han Pen
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Xiaoyan Song
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Jialei Chen
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Bo Xia
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Shunshun Han
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| |
Collapse
|
4
|
Yan X, Niu Y, Wang Y, Wei S, Han L, Guo Z, Zhao L, Gao F. CMSP exerts anti-tumor effects on small cell lung cancer cells by inducing mitochondrial dysfunction and ferroptosis. Open Med (Wars) 2025; 20:20241100. [PMID: 39822985 PMCID: PMC11737370 DOI: 10.1515/med-2024-1100] [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: 05/16/2024] [Revised: 10/09/2024] [Accepted: 11/07/2024] [Indexed: 01/19/2025] Open
Abstract
Purpose This study aims to investigate the role and mechanism of p-hydroxyl cinnamaldehyde (CMSP) in triggering ferroptosis of small cell lung cancer (SCLC) cells. Methods The impact of CMSP on ferroptosis in H1688 and SW1271 cells was assessed through cell experiments and biological information analysis. Moreover, the expression of heme oxygenase 1 (HMOX1) in SCLC tissue was examined. Results Following CMSP treatment, a concentration-dependent increase in cell death was observed, and differentially expressed genes were found to be associated with ferroptosis. CMSP notably facilitated ferroptosis events, such as elevated levels of reactive oxygen species (ROS), Fe2+, malondialdehyde (MDA), transferrin receptor 1 (TFR1), divalent metal transporter 1 (DMT1), and decreased levels of glutathione (GSH), solute carrier family 7 member 11 (SLC7A11), and glutathione peroxidase 4 (GPX4). Furthermore, CMSP promoted mitochondrial dysfunction, manifested as reduced mitochondrial volume, increased membrane density, elevated mitochondrial ROS, and decreased mitochondrial membrane potential. Consistently, the mitochondrial-targeted antioxidant Mito-TEMPO reversed CMSP-induced ferroptosis. Expression of the HMOX1 gene was markedly increased under CMSP treatment, while lower expression was observed in cancer tissue compared to adjacent tissue. Conclusion CMSP triggers mitochondrial dysfunction via HMOX1 activation, leading to ferroptosis in SCLC cells, underscoring its potential as a therapeutic agent for SCLC.
Collapse
Affiliation(s)
- Xi Yan
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yinghao Niu
- Department of Clinical Biobank, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, China
| | - Yaojie Wang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Sisi Wei
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Lina Han
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Zhongyu Guo
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Feng Gao
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| |
Collapse
|
5
|
Zhu Y, Kang N, Zhang L, Tao J, Xue W, Li H, Li Y, Zheng X, He W, Ma J. Targeting and degradation of OTUB1 by Erianin for antimetastasis in esophageal squamous cell carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:155969. [PMID: 39566402 DOI: 10.1016/j.phymed.2024.155969] [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: 01/18/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Metastasis is a major contributor to mortality in patients with esophageal squamous cell carcinoma (ESCC); effective treatment is currently lacking. Erianin, a bioactive ingredient of traditional Chinese medicine, Dendrobium chrysotoxum, has anti-tumor activity against multiple human tumors. However, the effect and associated underlying mechanism of Erianin on ESCC antimetastasis remain unclear. PURPOSE To investigate the anti-metastatic properties of Erianin in ESCC both in vitro and in vivo and associated molecular mechanisms. METHODS Wound healing assay, Transwell assay, CCK-8 assay, immunohistochemistry, and lung metastasis mouse model were carried out to examine ESCC cell migration and viability in vitro and in vivo. Drug affinity responsive target stability (DARTS), cellular thermal migration assay (CETSA), molecular docking, and Surface plasmon resonance (SPR) assay were used to confirm Erianin binding to ovarian tumor ubiquitin aldehyde-binding protein 1 (OTUB1) protein. Protein stability assay, cell transfection, and western blotting were used to confirm Erianin-mediated degradation of OTUB1 and Snail via the ubiquitin-proteasome pathway. qRT-PCR and western blotting were used to assess OTUB1expression in ESCC tissues. RESULTS Erianin suppressed the migration/invasion of ESCC cells without modulating cell viability in vitro and in vivo, bound to OTUB1 through DARTS, CETSA, and molecular docking, and SPR assay, and enhanced OTUB1 degradation via the ubiquitin-proteasome system. Moreover, Erianin inhibited the ESCC epithelial-mesenchymal transition by enhancing the ubiquitination and degradation of Snail via targeting OTUB1. CONCLUSION Erianin inhibited ESCC metastasis through ubiquitination and degradation of Snail via targeting OTUB1. Our findings suggest Erianin as a novel OTUB1 inhibitor for preventing ESCC metastasis.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Ningning Kang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, PR China
| | - Li Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Jianju Tao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Wen Xue
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Hui Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Yingcan Li
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Xucai Zheng
- Department of Head, Neck and Breast Surgery, the First Affiliated Hospital of USTC, Anhui Provincial Cancer Hospital, Hefei, Anhui 230031, PR China.
| | - Wei He
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China.
| | - Junting Ma
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China.
| |
Collapse
|
6
|
Zhu L, Du Y. A promising new approach to cancer therapy: Manipulate ferroptosis by hijacking endogenous iron. Int J Pharm 2024; 662:124517. [PMID: 39084581 DOI: 10.1016/j.ijpharm.2024.124517] [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: 04/08/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Ferroptosis, a form of regulated cell death characterized by iron-dependent phospholipid peroxidation, has emerged as a focal point in the field of cancer therapy. Compared with other cell death modes such as apoptosis and necrosis, ferroptosis exhibits many distinct characteristics in the molecular mechanisms and cell morphology, offering a promising avenue for combating cancers that are resistant to conventional therapeutic modalities. In light of the serious side effects associated with current Fenton-modulating ferroptosis therapies utilizing exogenous iron-based inorganic nanomaterials, hijacking endogenous iron could serve as an effective alternative strategy to trigger ferroptosis through targeting cellular iron regulatory mechanisms. A better understanding of the underlying iron regulatory mechanism in the process of ferroptosis has shed light on the current findings of endogenous ferroptosis-based nanomedicine strategies for cancer therapy. Here in this review article, we provide a comprehensive discussion on the regulatory network of iron metabolism and its pivotal role in ferroptosis, and present recent updates on the application of nanoparticles endowed with the ability to hijack endogenous iron for ferroptosis. We envision that the insights in the study may expedite the development and translation of endogenous ferroptosis-based nanomedicines for effective cancer treatment.
Collapse
Affiliation(s)
- Luwen Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang 321299, China.
| |
Collapse
|
7
|
Zheng X, Tang P, Li H, Ye T, Zhu X, He W, Cheng L, Cheng H. Cucurbitacin E elicits apoptosis in laryngeal squamous cell carcinoma by enhancing reactive oxygen species-regulated mitochondrial dysfunction and endoplasmic reticulum stress. Am J Cancer Res 2024; 14:3905-3921. [PMID: 39267666 PMCID: PMC11387858 DOI: 10.62347/hpqq9412] [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: 06/04/2024] [Accepted: 08/18/2024] [Indexed: 09/15/2024] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a prevalent head and neck neoplasm with escalating global morbidity and mortality rates. Despite the increasing burden of LSCC, the drugs currently approved for its treatment are limited. Therefore, it is necessary to identify novel and promising drugs that target LSCC. Cucurbitacin E (CuE) is a naturally oxygenated tetracyclic triterpenoid that suppresses several cancers. However, its anti-LSCC activity and the molecular mechanisms of action remain unclear. This study explored its impact on LSCC, revealing cell viability attenuation and apoptosis enhancement in vitro. Further investigations indicated that CuE significantly decreased mitochondrial membrane potential, thereby promoting cytochrome c release, increasing cleaved-Caspase 3 and cleaved-PARP levels, and triggering mitochondria-dependent apoptosis. Concurrently, exposure of LSCC cells to CuE enhanced endoplasmic reticulum (ER) stress, mobilized the protein kinase RNA-like endoplasmic reticulum kinase/initiation factor 2a/ATF4/C-EBP homologous protein pathway, and induced LSCC cell apoptosis. Finally, CuE markedly elevated intracellular reactive oxygen species (ROS) levels. When ROS were eliminated with N-acetylcysteine, CuE-mediated mitochondrial dysfunction, ER stress, and cell apoptosis were nearly abolished. Similar outcomes were observed in murine LSCC models. Together, these results highlight that CuE suppresses proliferation while triggering apoptosis in LSCC cells via ROS-regulated mitochondrial dysfunction and the ER stress pathway. Hence, CuE may serve as a promising candidate for LCSS treatment.
Collapse
Affiliation(s)
- Xucai Zheng
- Department of Oncology, The Second Hospital of Anhui Medical University Hefei 230601, Anhui, China
- Department of Head, Neck and Breast Surgery, The First Affiliated Hospital of USTC, Anhui Provincial Cancer Hospital Hefei 230031, Anhui, China
| | - Puze Tang
- Bachelor of Science in Mathematics, University of Liverpool United Kingdom
| | - Hui Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Tingbo Ye
- Department of Head, Neck and Breast Surgery, The First Affiliated Hospital of USTC, Anhui Provincial Cancer Hospital Hefei 230031, Anhui, China
| | - Xu Zhu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Wei He
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Ling Cheng
- Medical Intensive Care Unit, The First Affiliated Hospital of Anhui University of Chinese Medicine Hefei 230031, Anhui, China
| | - Huaidong Cheng
- Department of Oncology, The Second Hospital of Anhui Medical University Hefei 230601, Anhui, China
- Department of Oncology, Shenzhen Hospital of Southern Medical University Shenzhen 518000, Guangdong, China
| |
Collapse
|
8
|
Luo Y, Bai XY, Zhang L, Hu QQ, Zhang N, Cheng JZ, Hou MZ, Liu XL. Ferroptosis in Cancer Therapy: Mechanisms, Small Molecule Inducers, and Novel Approaches. Drug Des Devel Ther 2024; 18:2485-2529. [PMID: 38919962 PMCID: PMC11198730 DOI: 10.2147/dddt.s472178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Ferroptosis, a unique form of programmed cell death, is initiated by an excess of iron accumulation and lipid peroxidation-induced damage. There is a growing body of evidence indicating that ferroptosis plays a critical role in the advancement of tumors. The increased metabolic activity and higher iron levels in tumor cells make them particularly vulnerable to ferroptosis. As a result, the targeted induction of ferroptosis is becoming an increasingly promising approach for cancer treatment. This review offers an overview of the regulatory mechanisms of ferroptosis, delves into the mechanism of action of traditional small molecule ferroptosis inducers and their effects on various tumors. In addition, the latest progress in inducing ferroptosis using new means such as proteolysis-targeting chimeras (PROTACs), photodynamic therapy (PDT), sonodynamic therapy (SDT) and nanomaterials is summarized. Finally, this review discusses the challenges and opportunities in the development of ferroptosis-inducing agents, focusing on discovering new targets, improving selectivity, and reducing toxic and side effects.
Collapse
Affiliation(s)
- YiLin Luo
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Xin Yue Bai
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Lei Zhang
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Qian Qian Hu
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Ning Zhang
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Jun Zhi Cheng
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Ming Zheng Hou
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Xiao Long Liu
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| |
Collapse
|
9
|
Long D, Mao C, Huang Y, Xu Y, Zhu Y. Ferroptosis in ulcerative colitis: Potential mechanisms and promising therapeutic targets. Biomed Pharmacother 2024; 175:116722. [PMID: 38729051 DOI: 10.1016/j.biopha.2024.116722] [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: 03/05/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
Ulcerative colitis (UC) is a complex immune-mediated chronic inflammatory bowel disease. It is mainly characterized by diffuse inflammation of the colonic and rectal mucosa with barrier function impairment. Identifying new biomarkers for the development of more effective UC therapies remains a pressing task for current research. Ferroptosis is a newly identified form of regulated cell death characterized by iron-dependent lipid peroxidation. As research deepens, ferroptosis has been demonstrated to be involved in the pathological processes of numerous diseases. A growing body of evidence suggests that the pathogenesis of UC is associated with ferroptosis, and the regulation of ferroptosis provides new opportunities for UC treatment. However, the specific mechanisms by which ferroptosis participates in the development of UC remain to be more fully and thoroughly investigated. Therefore, in this review, we focus on the research advances in the mechanism of ferroptosis in recent years and describe the potential role of ferroptosis in the pathogenesis of UC. In addition, we explore the underlying role of the crosslinked pathway between ferroptosis and other mechanisms such as macrophages, neutrophils, autophagy, endoplasmic reticulum stress, and gut microbiota in UC. Finally, we also summarize the potential compounds that may act as ferroptosis inhibitors in UC in the future.
Collapse
Affiliation(s)
- Dan Long
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chenhan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yingtao Huang
- The First Clinical Medical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yin Xu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China.
| | - Ying Zhu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
10
|
Yu X, He Z, Wang Z, Ke S, Wang H, Wang Q, Li S. Brusatol hinders the progression of bladder cancer by Chac1/Nrf2/SLC7A11 pathway. Exp Cell Res 2024; 438:114053. [PMID: 38663476 DOI: 10.1016/j.yexcr.2024.114053] [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: 01/14/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Bladder cancer is a common tumor that impacts the urinary system and marked by a significant fatality rate and an unfavorable prognosis. Promising antineoplastic properties are exhibited by brusatol, which is obtained from the dried ripe fruit of Brucea javanica. The present study aimed to evaluate the influence of brusatol on the progression of bladder cancer and uncover the molecular mechanism involved. We used Cell Counting Kit-8, colony formation and EdU assays to detect cell numbers, viability and proliferation. We used transwell migration assay to detect cell migration ability. The mechanism of brusatol inhibition of bladder cancer proliferation was studied by flow cytometry and western blotting. It was revealed that brusatol could reduce the viability and proliferation of T24 and 5637 cells. The transwell migration assay revealed that brusatol was able to attenuate the migration of T24 and 5637 cells. We found that treatment with brusatol increased the levels of reactive oxygen species, malondialdehyde and Fe2+, thereby further promoting ferroptosis in T24 and 5637 cells. In addition, treatment with RSL3 (an agonistor of ferroptosis) ferrostatin-1 (a selective inhibitor of ferroptosis) enhanced or reversed the brusatol-induced inhibition. In vivo, treatment with brusatol significantly suppressed the tumor growth in nude mice. Mechanistically, brusatol induced ferroptosis by upregulating the expression of ChaC glutathione-specific gamma-glutamylcyclotransferase (Chac1) and decreasing the expression of SLC7A11 and Nrf2 in T24 and 5637 cells. To summarize, the findings of this research demonstrated that brusatol hindered the growth of bladder cancer and triggered ferroptosis via the Chac1/Nrf2/SLC7A11 pathway.
Collapse
Affiliation(s)
- Xi Yu
- Departments of Anesthesiology of Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Ziqi He
- Departments of Anesthesiology of Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Departments of urology of Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhong Wang
- Departments of Anesthesiology of Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei 430060, PR China.
| | - Shuai Ke
- Departments of Anesthesiology of Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Departments of urology of Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Huaxin Wang
- Departments of Anesthesiology of Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qinghua Wang
- Departments of Anesthesiology of Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Department of Urology, Tongji Hospital, Tongji University School of Medicine, 200065 Shanghai, China
| | - Shenglan Li
- Departments of Anesthesiology of Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Department of Radiography, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei, 430060, PR China.
| |
Collapse
|
11
|
Yang YY, An YC, Zhang SY, Huang MY, Ye XQ, Zhao ZH, Liu WC. Biogenic Selenium Nanoparticles Synthesized Using Alginate Oligosaccharides Attenuate Heat Stress-Induced Impairment of Breast Meat Quality via Regulating Oxidative Stress, Metabolome and Ferroptosis in Broilers. Antioxidants (Basel) 2023; 12:2032. [PMID: 38136152 PMCID: PMC10740886 DOI: 10.3390/antiox12122032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Selenium (Se) is an indispensable trace element with versatile functions in antioxidant defense in poultry. In our previous study, we synthesized a novel type of biogenic selenium nanoparticle based on alginate oligosaccharides (SeNPs-AOS), and found that the particles are sized around 80 nm with an 8% Se content, and the dietary addition of 5 mg/kg of SeNPs-AOS could effectively alleviate the deleterious effects of heat stress (HS) in broilers, but it is still unclear whether SeNPs-AOS can improve the meat quality. Therefore, the aim of this study was to evaluate the protective effects of SeNPs-AOS on breast meat quality in heat-stressed broilers, and explore the relevant mechanisms. Birds at the age of 21 days were randomly divided into four groups with six replicates per group (eight broilers per replicate) according to a 2 × 2 experimental design, using HS (33 ± 2 °C, 10 h/day vs. thermoneutral, TN, under 23 ± 1.5 °C) and SeNPs-AOS (5 mg/kg feed vs. no inclusion) as variables. The results showed that dietary SeNPs-AOS decreased the cooking loss (p < 0.05), freezing loss (p < 0.001), and shear force (p < 0.01) of breast muscle in heat-stressed broilers. The non-targeted metabolomics analysis of the breast muscle identified 78 differential metabolites between the HS and HS + SeNPs-AOS groups, mainly enriched in the arginine and proline metabolism, β-alanine metabolism, D-arginine and D-ornithine metabolism, pantothenate, and CoA biosynthesis pathways (p < 0.05). Meanwhile, supplementation with SeNPs-AOS increased the levels of the total antioxidant capacity (T-AOC), the activities of catalase (CAT) and glutathione peroxidase (GSH-Px), and decreased the content of malondialdehyde (MDA) in the breast muscle (p < 0.05) in broilers under HS exposure. Additionally, SeNPs-AOS upregulated the mRNA expression of CAT, GPX1, GPX3, heme oxygenase-1 (HO-1), masculoaponeurotic fibrosarcoma G (MafG), MafK, selenoprotein W (SELENOW), SELENOK, ferritin heavy polypeptide-1 (FTH1), Ferroportin 1 (Fpn1), and nuclear factor erythroid 2-related factor 2 (Nrf2) (p < 0.05), while it downregulated Kelch-like ECH-associated pro-36 tein 1 (Keap1) and prostaglandin-endoperoxide Synthase 2 (PTGS2) expression (p < 0.05) in broilers under HS. These findings demonstrated that the dietary addition of SeNPs-AOS mitigated HS-induced oxidative damage and metabolite changes in the breast muscle of broilers, which may be related to the regulation of the Nrf2 signaling pathway and selenoprotein synthesis. In addition, SeNPs-AOS upregulated the breast muscle gene expression of anti-ferroptosis-related molecules in broilers under HS, suggesting that SeNPs-AOS can be used as novel Se supplements against HS in broilers.
Collapse
Affiliation(s)
- Yu-Ying Yang
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Y.-Y.Y.); (S.-Y.Z.); (M.-Y.H.); (X.-Q.Y.)
| | - Yu-Chen An
- School of Computer Science and Engineering, Yangjiang Campus, Guangdong Ocean University, Yangjiang 529500, China;
| | - Shu-Yue Zhang
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Y.-Y.Y.); (S.-Y.Z.); (M.-Y.H.); (X.-Q.Y.)
| | - Meng-Yi Huang
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Y.-Y.Y.); (S.-Y.Z.); (M.-Y.H.); (X.-Q.Y.)
| | - Xue-Qing Ye
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Y.-Y.Y.); (S.-Y.Z.); (M.-Y.H.); (X.-Q.Y.)
| | - Zhi-Hui Zhao
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Y.-Y.Y.); (S.-Y.Z.); (M.-Y.H.); (X.-Q.Y.)
| | - Wen-Chao Liu
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Y.-Y.Y.); (S.-Y.Z.); (M.-Y.H.); (X.-Q.Y.)
| |
Collapse
|