1
|
Peng X, Feng J, Yang H, Xia P, Pu F. Nrf2: A key regulator in chemoradiotherapy resistance of osteosarcoma. Genes Dis 2025; 12:101335. [PMID: 40242036 PMCID: PMC12000747 DOI: 10.1016/j.gendis.2024.101335] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/24/2024] [Accepted: 04/03/2024] [Indexed: 04/18/2025] Open
Abstract
Osteosarcoma (OS), frequently observed in children and adolescents, is one of the most common primary malignant tumors of the bone known to be associated with a high capacity for invasion and metastasis. The incidence of osteosarcoma in children and adolescents is growing annually, although improvements in survival remain limited. With the clinical application of neoadjuvant chemotherapy, chemotherapy combined with limb-preserving surgery has gained momentum as a major intervention. However, certain patients with OS experience treatment failure owing to chemoradiotherapy resistance or metastasis. Nuclear factor E2-related factor 2 (Nrf2), a key antioxidant factor in organisms, plays a crucial role in maintaining cellular physiological homeostasis; however, its overactivation in cancer cells restricts reactive oxygen species production, promotes DNA repair and drug efflux, and ultimately leads to chemoradiotherapy resistance. Recent studies have also identified the functions of Nrf2 beyond its antioxidative function, including the promotion of proliferation, metastasis, and regulation of metabolism. The current review describes the multiple mechanisms of chemoradiotherapy resistance in OS and the substantial role of Nrf2 in the signaling regulatory network to elucidate the function of Nrf2 in promoting OS chemoradiotherapy resistance and formulating relevant therapeutic strategies.
Collapse
Affiliation(s)
- Xianglin Peng
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| | - Jing Feng
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| | - Han Yang
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Ping Xia
- Department of Orthopedics, Wuhan Fourth Hospital, Wuhan 430030, China
| | - Feifei Pu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| |
Collapse
|
2
|
Tang K, Ye T, He Y, Ba X, Xia D, Peng E, Chen Z, Ye Z, Yang X. Ferroptosis, necroptosis, and pyroptosis in calcium oxalate crystal-induced kidney injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167791. [PMID: 40086520 DOI: 10.1016/j.bbadis.2025.167791] [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: 11/30/2024] [Revised: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Kidney stones represent a highly prevalent urological disorder worldwide, with high incidence and recurrence rates. Calcium oxalate (CaOx) crystal-induced kidney injury serves as the foundational mechanism for the formation and progression of CaOx stones. Regulated cell death (RCD) such as ferroptosis, necroptosis, and pyroptosis are essential in the pathophysiological process of kidney injury. Ferroptosis, a newly discovered RCD, is characterized by its reliance on iron-mediated lipid peroxidation. Necroptosis, a widely studied programmed necrosis, initiates with a necrotic phenotype that resembles apoptosis in appearance. Pyroptosis, a type of RCD that involves the gasdermin protein, is accompanied by inflammation and immune response. In recent years, increasing amounts of evidence has demonstrated that ferroptosis, necroptosis, and pyroptosis are significant pathophysiological processes involved in CaOx crystal-induced kidney injury. Herein, we summed up the roles of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury. Furthermore, we delved into the curative potential of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury.
Collapse
Affiliation(s)
- Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Ye
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Li ZT, Lin T, Sun Y, Wang XY, Yang YX, Gan L, Xu JM, Wei XT, Zhu HQ, Zhao WC, Zhu ZH. Expression and purification of recombinant glutaredoxin 1 and protection against oxidative stress injury during cerebral ischemia-reperfusion injury. Protein Expr Purif 2025; 230:106689. [PMID: 39971172 DOI: 10.1016/j.pep.2025.106689] [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: 12/13/2024] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Glutaredoxin (Grx) is a small molecular protein widely found in both prokaryotes and eukaryotes, serving various biological functions, including participation in redox reactions and exerting anti-apoptotic effects[1]. To evaluate the protective effect of recombinant Grx1 against oxidative stress, we constructed the pET-30a (+)/Grx1 recombinant plasmid and performed soluble expression and purification of the recombinant Grx1. In vitro experiments, including ABTS and DPPH radical scavenging assays, showed that recombinant Grx1 has significant antioxidant activity. Reactive oxygen species detection revealed that the levels of reactive oxygen species in the Grx1 treatment group decreased by 33.01 % compared to the H2O2 group. Flow cytometry analyses indicated that the number of apoptotic cells in the Grx1 treatment group decreased by 23.51 % relative to the H2O2 group. Additionally, qRT-PCR analysis showed that Grx1 significantly reduced the expression levels of genes such as IL-1β, TNF-α, IL-6, and caspase-3 in PC12 cells. In vivo, recombinant Grx1 was utilized to treat cerebral ischemia-reperfusion injury (CIRI). Histological staining revealed that recombinant Grx1 significantly mitigated hippocampal tissue damage. Western blotting analysis demonstrated that Grx1 can reduce neuronal apoptosis following CIRI by decreasing Bax expression while increasing Bcl-2 expression. Furthermore, Grx1 was shown to modulate the HO-1/Nrf2 signaling pathway by elevating the expression of Nrf2 and HO-1. In summary, this study successfully overexpressed biologically active Grx1 in E. coli, and confirms that recombinant Grx1 exhibits remarkable antioxidant activity in both in vitro and in vivo experiments, effectively alleviating oxidative stress damage associated with ischemic stroke.
Collapse
Affiliation(s)
- Zi-Teng Li
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Tong Lin
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Yu Sun
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Xin-Yi Wang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Yi-Xuan Yang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Li Gan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Jia-Ming Xu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Xu-Ting Wei
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Huang-Qing Zhu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Wei-Chun Zhao
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China.
| | - Zhen-Hong Zhu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China.
| |
Collapse
|
4
|
Zhang H, Zhang D, Wang H, Liu Y, Ding W, Fan G, Meng X. Heme oxygenase 1‑overexpressing bone marrow mesenchymal stem cell‑derived exosomes suppress interleukin‑1 beta‑induced apoptosis and aging of nucleus pulposus cells. Mol Med Rep 2025; 31:116. [PMID: 40052562 PMCID: PMC11905203 DOI: 10.3892/mmr.2025.13481] [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: 09/23/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Exosomes derived from bone marrow mesenchymal stem cells (BMSCs) and heme oxygenase 1 (HO‑1) attenuate intervertebral disc degeneration (IVDD). However, whether BMSC‑derived exosomes attenuate IVDD by delivering HO‑1 to nucleus pulposus (NP) cells remains to be elucidated. Mouse BMSCs were characterized by multilineage differentiation and surface marker molecule detection. Exosomes Exo and Exo‑HO‑1 were isolated from BMSCs and HO‑1‑overexpressing BMSCs by ultracentrifugation and characterized by observing their morphology, detecting the exosome marker proteins, tumor susceptibility gene 101 (TSG101) and CD63 and analyzing their particle size. Interleukin‑1 β (IL‑1β)‑stimulated NP cells were used as the IVDD cell model. The influence of Exo or Exo‑HO‑1 on IL‑1β‑urged apoptosis and senescence in NP cells was determined by flow cytometry, western blotting and senescence‑associated β‑galactosidase (SA‑β‑gal) staining. Exo and Exo‑HO‑1 did not vary in size or morphology. Exo‑HO‑1 markedly repressed IL‑1β‑prompted apoptosis in NP cells, accompanied with a prominent increase in Cleaved caspase 3 and Bax protein levels and a marked decrease in Bcl‑2 protein levels. Exo and Exo‑HO‑1 both decreased the number of SA‑β‑gal‑positive NP cells and arrested NP cells in the G1 phase. Exo‑HO‑1 had stronger effects than Exo, suggesting that Exo‑HO‑1 can weaken IL‑1β‑induced NP cell senescence. In addition, Exo and Exo‑HO‑1 repressed IL‑1β mediating the phosphorylation of p65 and nuclear translocation of p65. In conclusion, HO‑1‑overexpressing BMSC‑derived exosomes blocked the nuclear factor‑kappa B signaling in IL‑1β‑stimulated NP cells, thus impairing cell apoptosis and senescence.
Collapse
Affiliation(s)
- Hao Zhang
- Spinal Surgery Department 2, Hebei Medical University Third Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Di Zhang
- Spinal Surgery Department 2, Hebei Medical University Third Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Hui Wang
- Spinal Surgery Department 2, Hebei Medical University Third Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Yilei Liu
- Spinal Surgery Department 2, Hebei Medical University Third Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Wenyuan Ding
- Spinal Surgery Department 2, Hebei Medical University Third Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Guangpu Fan
- Department of Cardiac Surgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Xianzhong Meng
- Spinal Surgery Department 1, Hebei Medical University Third Hospital, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
5
|
Wu B, Li D, Wang Y, Pan T, Xu J, Li L. The m6A methyltransferase METTL3 affects ferroptosis in non-small cell lung cancer by regulating the PTEN/PI3K/AKT pathway. Discov Oncol 2025; 16:559. [PMID: 40249573 PMCID: PMC12008084 DOI: 10.1007/s12672-025-02330-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) poses a major threat to human health, METTL3 has been reported to promote numerous tumor development by inhibiting ferroptosis. The aim of the present study was to explore the mechanism of action of METTL3 in NSCLC. METHODS The UALCAN online platform was applied to analyze METTL3 and PTEN expression in NSCLC and their relationship with tumor stages. NCI-H23 and NCI-H1975 cells were transfected with sh-METTL3, or oe-METTL3 respectively. Then EdU assay was employed to assess cell proliferation and the transwell assay was employed to assess the ability of cells to migrate and invade. Apoptosis was detected using flow cytometry. In addition, m6A methylation levels, oxidative stress indicators, and Fe2+ content were determined. Furthermore, GPX4 and PTEN expression, as well as PI3K and AKT phosphorylation were quantified. Finally, the cells with METTL3 knockdown were further transfected with sh-PTEN. RESULTS METTL3 expression was up-regulated in NSCLC and was closely related to the tumor stages. METTL3 overexpression significantly promoted the malignant phenotype of NSCLC cells, increased the methylation level of m6A mRNA, reduced oxidative stress, inhibited the occurrence of ferroptosis and apoptosis, and led to increased expression of GPX4 and activation of the PTEN/PI3K/AKT pathway. Conversely, METTL3 knockdown produced the opposite effect. Importantly, METTL3 knockdown-induced oxidative stress and ferroptosis in NCI-H23 cells were rescued by sh-PTEN or ferroptosis inhibitor Ferrostatin-1. CONCLUSION METTL3 may inhibit ferroptosis in NSCLC by activating the PTEN/PI3K/AKT pathway, suggesting that METTL3-mediated PTEN/PI3K/AKT pathway may be a promising therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Bin Wu
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China.
| | - Danhong Li
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China.
| | - Yu Wang
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Tingting Pan
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Jieru Xu
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Lihong Li
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| |
Collapse
|
6
|
Huang M, Wu Y, Wei X, Cheng L, Fu L, Yan H, Wei W, Li B, Ru H, Mo X, Tang W, Su Z, Yan L. Trifluridine/tipiracil induces ferroptosis by targeting p53 via the p53-SLC7A11 axis in colorectal cancer 3D organoids. Cell Death Dis 2025; 16:255. [PMID: 40188162 PMCID: PMC11972347 DOI: 10.1038/s41419-025-07541-z] [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: 09/19/2024] [Revised: 02/15/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
Trifluridine/Tipiracil (FTD/TPI, TAS102) has been approved for the treatment of patients with colorectal cancer (CRC) for its promising anticancer activity enabled by its incorporation into double strands during DNA synthesis. However, the mechanisms underlying the anticancer targets of FTD/TPI remain not fully understood. Here we report our observation of the activation of ferroptosis in CRC by FTD/TPI. Mechanistically, FTD/TPI directly promotes the ubiquitination and degradation of MDM2, thereby stabilizing the p53. Nuclear accumulation of p53 subsequently downregulates SLC7A11 expression, leading to ferroptosis. Furthermore, we observed that FTD/TPI combined with sulfasalazine (SAS), a system Xc- inhibitor, works in a synergistic manner to induce ferroptosis and further inhibit the proliferation of CRC cells. Finally, we confirmed the synergistic effect of SAS and FTD/TPI on patient-derived organoids in vitro and patient-derived xenograft mouse models in vivo. Our findings are the first to reveal that FTD/TPI induces ferroptosis via the p53-SLC7A11 axis and that SAS enhances the sensitivity and therapeutic effect of FTD/TPI. These findings suggest that the synergistic effect of FTD/TPI and SAS may represent a new therapeutic strategy for patients with CRC.
Collapse
Affiliation(s)
- Maosen Huang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Yancen Wu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xiaoxia Wei
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Linyao Cheng
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Lihua Fu
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Haochao Yan
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Wene Wei
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Bo Li
- Liaoning Provincial Engineering Laboratory of Anti-tumor Immunity and Molecular Theranostics Technology, Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Haiming Ru
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xianwei Mo
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Weizhong Tang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Zijie Su
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| | - Linhai Yan
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
7
|
Ye Z, Cheng L, Xuan Y, Yu K, Li J, Gu H. Chlorogenic acid alleviates the development of severe acute pancreatitis by inhibiting NLPR3 Inflammasome activation via Nrf2/HO-1 signaling. Int Immunopharmacol 2025; 151:114335. [PMID: 39987635 DOI: 10.1016/j.intimp.2025.114335] [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: 12/28/2024] [Revised: 02/15/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Severe acute pancreatitis (SAP), marked by profound tissue inflammation within the pancreatic tissue, is an abrupt and intense inflammation of the pancreas. Chlorogenic acid (CGA) is one of the effective pharmacological ingredients components in JinHong Tablet (JHT). The role of CGA in protecting pancreas from severe injury in pancreatitis needs to be studied. The intervention with CGA led to a significant decline in serum amylase and lipase levels in rats with SAP, concurrently mitigating the pathological impairment within the pancreatic tissue. CGA effectively diminishes the levels of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) in SAP rats by inhibiting the activation of NF-κB and the NLRP3 inflammasome. Additionally, in AR42J cells, the application of CGA was found to reduce the inflammatory response induced by caerulein. Mechanically, CGA alleviates the inflammatory response in SAP models by activating the Nrf2/HO-1 pathway. Together, CGA reduces the inflammatory response of SAP by activating the Nrf2/HO-1 pathway, thus alleviating the development of SAP. Our results provide a basis for the treatment of SAP.
Collapse
Affiliation(s)
- Zhen Ye
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China
| | - Lin Cheng
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China
| | - Yujun Xuan
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China
| | - Kui Yu
- Department of General Surgery, Pudong Branch, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiong Li
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China.
| | - Honggang Gu
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China.
| |
Collapse
|
8
|
Ouyang X, Wang J, Qiu X, Hu D, Cui J. Current developments of pharmacotherapy targeting heme oxygenase 1 in cancer (Review). Int J Oncol 2025; 66:26. [PMID: 39981901 DOI: 10.3892/ijo.2025.5732] [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: 11/25/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
Malignant tumors are non-communicable diseases that impact human health and quality of life. Identifying and targeting the underlying genetic drivers is a challenge. Heme oxygenase-1 (HO-1), a stress-inducible enzyme also known as heat shock protein 32, plays a crucial role in maintaining cellular homeostasis. It mitigates oxidative stress-induced damage and exhibits anti-apoptotic properties. HO-1 is expressed in a wide range of malignancies and is associated with tumor growth. However, the precise role of HO-1 in tumor development remains controversial. Drugs, both naturally occurring and chemically synthesized, can inhibit tumor growth by modulating HO-1 expression in cancer cells. The present review aimed to discuss biological functions of HO-1 pharmacological therapies targeting HO-1.
Collapse
Affiliation(s)
- Xiaohu Ouyang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jingbo Wang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaoyuan Qiu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jing Cui
- Health Management Center, Hubei Provincial Hospital of Integrated Chinese & Western Medicine, Wuhan, Hubei 430015, P.R. China
| |
Collapse
|
9
|
Wang Y, Li N, Guan W, Wang D. Controversy and multiple roles of the solitary nucleus receptor Nur77 in disease and physiology. FASEB J 2025; 39:e70468. [PMID: 40079203 PMCID: PMC11904867 DOI: 10.1096/fj.202402775rr] [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: 11/08/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Neuron-derived clone 77 (Nur77), a member of the orphan nuclear receptor family, is expressed and activated rapidly in response to diverse physiological and pathological stimuli. It exerts complex biological functions, including roles in the nervous system, genome integrity, cell differentiation, homeostasis, oxidative stress, autophagy, aging, and infection. Recent studies suggest that Nur77 agonists alleviate symptoms of neurodegenerative diseases, highlighting its potential as a therapeutic target in such conditions. In cancer, Nur77 demonstrates dual roles, acting as both a tumor suppressor and promoter, depending on the cancer type and stage, making it a controversial yet promising anticancer target. This review provides a structured analysis of the functions of Nur77, focusing on its physiological and pathological roles, therapeutic potential, and existing controversies. Emphasis is placed on its emerging applications in neurodegenerative diseases and cancer, offering key insights for future research and clinical translation.
Collapse
Affiliation(s)
- Yanteng Wang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Li
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wenwei Guan
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Difei Wang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
10
|
Li C, Deng D, Jiang Q, Shi J, Xu L, Liu Y. Ferroptosis in NAFLD: insights and the therapeutic potential of exercise. Front Med (Lausanne) 2025; 12:1462145. [PMID: 40206477 PMCID: PMC11979233 DOI: 10.3389/fmed.2025.1462145] [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: 07/09/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Ferroptosis, a distinct form of non-apoptotic cell death driven by iron accumulation, has garnered significant attention in recent years. Emerging evidence suggests that ferroptosis in hepatocytes may serve as a pivotal trigger in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Importantly, inhibiting ferroptosis has shown promising potential in slowing the progression of NAFLD. Concurrently, exercise, a cornerstone in the prevention and management of chronic diseases, plays a critical role in regulating disease progression. As such, the modulation of ferroptosis through exercise represents a promising avenue for developing innovative therapeutic strategies. This review aims to systematically elucidate the conceptual framework and molecular mechanisms underlying ferroptosis, with particular emphasis on its pathophysiological role in NAFLD. We have systematically summarized the effects of exercise on ferroptosis regulation through multiple molecular mechanisms, including upregulation of antioxidant defense systems via activation of NRF2, GPX4, and SLC7A11 signaling pathways; and modulation of iron metabolism through FPN-mediated iron homeostasis regulation. These findings not only provide valuable insights into the molecular basis of exercise-induced protection against ferroptosis-mediated cellular damage but also offer novel perspectives for future investigations into exercise-based interventions for NAFLD management. This work thereby contributes to the advancement of therapeutic strategies in the field of metabolic liver diseases.
Collapse
Affiliation(s)
- Chang Li
- Graduate School, Harbin Sport University, Harbin, Heilongjiang, China
| | - Dongkun Deng
- Graduate School, Harbin Sport University, Harbin, Heilongjiang, China
| | - Qingfeng Jiang
- Graduate School, Harbin Sport University, Harbin, Heilongjiang, China
| | - Jiaming Shi
- Graduate School, Harbin Sport University, Harbin, Heilongjiang, China
| | - Lin Xu
- College of Human Sport Science, Harbin Sport University, Harbin, Heilongjiang, China
| | - Yufei Liu
- College of Human Sport Science, Harbin Sport University, Harbin, Heilongjiang, China
| |
Collapse
|
11
|
Liu K, Yu J, Huang X, Gao H, Wang J. WGCNA and ferroptosis genes in OSCC: unraveling prognostic biomarkers and therapeutic targets. Discov Oncol 2025; 16:379. [PMID: 40126728 PMCID: PMC11933588 DOI: 10.1007/s12672-025-02151-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is the predominant type of oral cancer, with over 370,000 new cases and approximately 170,000 deaths annually worldwide. Despite therapeutic advancements, OSCC mortality rates have been increasing, underscoring the need for improved prognostic models and therapeutic targets. METHODS We integrated transcriptomic and clinical survival data from the TCGA-OSCC dataset to identify ferroptosis-related prognostic genes. Using weighted gene co-expression network analysis (WGCNA), we selected genes associated with OSCC prognosis and applied Lasso regression analysis to pinpoint key genes. A prognostic model was constructed and validated through survival analysis and receiver operating characteristic (ROC) curve analysis. RESULTS WGCNA identified modules significantly correlated with ferroptosis, yielding 321 genes associated with OSCC prognosis. Univariate Cox analysis identified 13 genes affecting OSCC prognosis. Lasso regression and multivariate Cox regression narrowed down the gene set to a final set of 7 genes, which were used to construct the risk model. The model stratified patients into high- and low-risk groups with significant survival differences (P < 0.001). The model's predictive accuracy was validated, with AUC values ranging from 0.565 to 0.733 for 1-, 3-, and 5-year survival predictions. Immune-related analysis revealed that low-risk patients exhibited higher immune cell infiltration and were more likely to benefit from immunotherapy. CONCLUSION Our study presents a novel prognostic model for OSCC patients based on ferroptosis-related genes, which not only predicts survival but also identifies potential therapeutic targets. The model's predictive accuracy and clinical relevance were validated, offering a new strategy for OSCC treatment.
Collapse
Affiliation(s)
- Ke Liu
- Department of Stomatology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Jiannan Yu
- Department of Pediatric Dental Prevention, The Afiliated Stomatological Hospial of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xuanxi Huang
- The Ninth Outpatient Department, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hongyan Gao
- Department of Health Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jing Wang
- Department of Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
12
|
Teng C, Chen JW, Shen LS, Chen S, Chen GQ. Research advances in natural sesquiterpene lactones: overcoming cancer drug resistance through modulation of key signaling pathways. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:13. [PMID: 40201307 PMCID: PMC11977367 DOI: 10.20517/cdr.2024.178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 04/10/2025]
Abstract
Cancer remains a significant global health challenge, with current chemotherapeutic strategies frequently limited by the emergence of resistance. In this context, natural compounds with the potential to overcome resistance have garnered considerable attention. Among these, sesquiterpene lactones, primarily derived from plants in the Asteraceae family, stand out for their potential anticancer properties. This review specifically focuses on five key signaling pathways: PI3K/Akt/mTOR, NF-κB, Wnt/β-catenin, MAPK/ERK, and STAT3, which play central roles in the mechanisms of cancer resistance. For each of these pathways, we detail their involvement in both cancer development and the emergence of drug resistance. Additionally, we investigate how sesquiterpene lactones modulate these pathways to overcome resistance across diverse cancer types. These insights highlight the potential of sesquiterpene lactones to drive the advancement of novel therapies that can effectively combat both cancer progression and drug resistance.
Collapse
Affiliation(s)
- Chi Teng
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, Guangdong, China
- Authors contributed equally
| | - Jia-Wen Chen
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, Guangdong, China
- Authors contributed equally
| | - Li-Sha Shen
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Sibao Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, Guangdong, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Guo-Qing Chen
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, Guangdong, China
| |
Collapse
|
13
|
Wang S, Song X, Gao H, Zhang Y, Zhou X, Wang F. 6-Gingerol Inhibits Ferroptosis in Endothelial Cells in Atherosclerosis by Activating the NRF2/HO-1 Pathway. Appl Biochem Biotechnol 2025:10.1007/s12010-025-05214-3. [PMID: 40120045 DOI: 10.1007/s12010-025-05214-3] [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] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Targeting endothelial cell ferroptosis is a potential approach for the treatment of atherosclerosis (AS). 6-Gingerol (6-Gin) is an active substance in ginger that is beneficial for improving AS. We conducted this study to explore whether 6-Gin mediated AS progression by regulating ferroptosis of endothelial cells. ApoE-/- mice were fed a high-fat diet to establish AS mouse model. Additionally, oxidized low-density lipoprotein (ox-LDL) was used to treat human umbilical vein endothelial cells (HUVECs) to generate injured cell model. Ferroptosis was evaluated by propidium iodide staining assay, western blot, and detecting iron, glutathione, malonaldehyde, and reactive oxygen species levels. The results showed that ox-LDL inhibited the proliferation and induced inflammation and ferroptosis of HUVECs, which was reversed by 6-Gin treatment. Moreover, 6-Gin upregulated HO-1 and NQO1 levels and promoted nuclear translocation of NRF2 in ox-LDL-treated HUVECs. ATRA, an NRF2 inhibitor, abrogated the promotion of proliferation and the inhibition of inflammation and ferroptosis induced by 6-Gin. Additionally, 6-Gin alleviated AS and suppressed ferroptosis in vivo. In conclusion, 6-Gin inhibited endothelial cell ferroptosis by inactivating the NRF2/HO-1 pathway, thereby improving abnormal lipid metabolism in AS mice. These findings suggest that 6-Gin may be a novel therapeutic drug for AS.
Collapse
Affiliation(s)
- Shuai Wang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xiaoliang Song
- Department of Geriatrics, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Hui Gao
- The First Clinical College of Liaoning, University of Traditional Chinese Medicine, Shenyang, China
| | - Yi Zhang
- The First Clinical College of Liaoning, University of Traditional Chinese Medicine, Shenyang, China
| | - Xin Zhou
- The First Clinical College of Liaoning, University of Traditional Chinese Medicine, Shenyang, China
| | - Fengrong Wang
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China.
| |
Collapse
|
14
|
Zhou X, Tian L, Xiong W, Li Y, Liu Q. Ferroptosis and hyperoxic lung injury: insights into pathophysiology and treatment approaches. Front Pharmacol 2025; 16:1568246. [PMID: 40170719 PMCID: PMC11958998 DOI: 10.3389/fphar.2025.1568246] [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: 01/29/2025] [Accepted: 03/04/2025] [Indexed: 04/03/2025] Open
Abstract
Hyperoxia therapy is a critical clinical intervention for both acute and chronic illnesses. However, prolonged exposure to high-concentration oxygen can cause lung injury. The mechanisms of hyperoxic lung injury (HLI) remain incompletely understood, and current treatment options are limited. Improving the safety of hyperoxia therapy has thus become an urgent priority. Ferroptosis, a novel form of regulated cell death characterized by iron accumulation and excessive lipid peroxidation, has been implicated in the pathogenesis of HLI, including diffuse alveolar damage, vascular endothelial injury, and bronchopulmonary dysplasia. In this review, we analyze the latest findings on ferroptosis and therapeutic strategies for HLI. Our aim is to provide new insights for the treatment of HLI and to facilitate the translation of these findings from bench to bedside.
Collapse
Affiliation(s)
- Xiaoqiong Zhou
- Department of Anesthesiology, Zigong First People’s Hospital, Zigong Academy of Medical Sciences, Zigong, China
| | - Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Wenyan Xiong
- Department of Anesthesiology, Yibin Maternity and Children Hospital, Yibin, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People’s Hospital, Zigong Academy of Medical Sciences, Zigong, China
| |
Collapse
|
15
|
Luo YY, Ba XY, Wang L, Zhang YP, Xu H, Chen PQ, Zhang LB, Han J, Luo H. LEF1 influences diabetic retinopathy and retinal pigment epithelial cell ferroptosis via the miR-495-3p/GRP78 axis through lnc-MGC. World J Diabetes 2025; 16:92003. [PMID: 40093269 PMCID: PMC11885969 DOI: 10.4239/wjd.v16.i3.92003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 11/10/2024] [Accepted: 12/11/2024] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is one of the major eye diseases contributing to blindness worldwide. Endoplasmic reticulum (ER) stress in retinal cells is a key factor leading to retinal inflammation and vascular leakage in DR, but its mechanism is still unclear. AIM To investigate the potential mechanism of LEF1 and related RNAs in DR. METHODS ARPE-19 cells were exposed to high levels of glucose for 24 hours to simulate a diabetic environment. Intraperitoneally injected streptozotocin was used to induce the rat model of DR. The expression levels of genes and related proteins were measured by RT-qPCR and Western blotting; lnc-MGC and miR-495-3p were detected by fluorescent in situ hybridization; CCK-8 and TUNEL assays were used to detect cell viability and apoptosis; enzyme-linked immunosorbent assay was used to detect inflammatory factors; dual-luciferase gene assays were used to verify the targeting relationship; and the retina was observed by HE staining. RESULTS LEF1 and lnc-MGC have binding sites, and lnc-MGC can regulate the miR-495-3p/GRP78 molecular axis. In high glucose-treated cells, inflammation was aggravated, the intracellular reactive oxygen species concentration was increased, cell viability was reduced, apoptosis was increased, the ER response was intensified, and ferroptosis was increased. As an ER molecular chaperone, GRP78 regulates the ER and ferroptosis under the targeting of miR-495-3p, whereas inhibiting LEF1 can further downregulate the expression of lnc-MGC, increase the level of miR-495-3p, and sequentially regulate the level of GRP78 to alleviate the occurrence and development of DR. Animal experiments indicated that the knockdown of LEF1 can affect the lnc-MGC/miR-495-3p/GRP78 signaling axis to restrain the progression of DR. CONCLUSION LEF1 knockdown can regulate the miR-495-3p/GRP78 molecular axis through lnc-MGC, which affects ER stress and restrains the progression of DR and ferroptosis in retinal pigment epithelial cells.
Collapse
Affiliation(s)
- Yi-Yi Luo
- Precision Medicine Center of Chuxiong Yi Autonomous Prefecture, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
| | - Xue-Ying Ba
- Precision Medicine Center of Chuxiong Yi Autonomous Prefecture, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
| | - Ling Wang
- Department of Endocrinology, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
| | - Ye-Pin Zhang
- Department of Pathology, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
| | - Hong Xu
- Department of Ophthalmology, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
| | - Pei-Qi Chen
- Department of Endocrinology, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
| | - Li-Bo Zhang
- Department of Ophthalmology, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
| | - Jian Han
- Precision Medicine Center of Chuxiong Yi Autonomous Prefecture, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
| | - Heng Luo
- Precision Medicine Center of Chuxiong Yi Autonomous Prefecture, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
- Department of Ophthalmology, The People's Hospital of Chuxiong Yi Autonomous Prefecture & The Fourth Affiliated Hospital of DaLi University, Chuxiong 675000, Yunnan Province, China
| |
Collapse
|
16
|
Shi M, Li X, Guo Y, Zhang Y, Xu J, Yan L, Liu R, Wang H, Tang S, Zhao Y, Li Z, Feng Y, Ren D, Liu P. Gaudichaudione H Enhances the Sensitivity of Hepatocellular Carcinoma Cells to Disulfidptosis via Regulating NRF2-SLC7A11 Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411131. [PMID: 39840687 PMCID: PMC11923960 DOI: 10.1002/advs.202411131] [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: 09/11/2024] [Revised: 12/01/2024] [Indexed: 01/23/2025]
Abstract
Gaudichaudione H (GH) is a naturally occurring small molecular compound derived from Garcinia oligantha Merr. (Clusiaceae), but the full pharmacological functions remain unclear. Herein, the potential of GH in disulfidptosis regulation, a novel form of programmed cell death induced by disulfide stress is explored. The omics results indicated that NRF2 signaling could be significantly activated by GH. The potential targets are associated with hepatocarcinogenesis and cell death. Moreover, both glutathione (GSH) metabolism and NADP+-NADPH metabolism are affected by GH, indicating the potential in disulfidptosis regulation. It is also observed that GH enhanced the sensitivity of hepatocellular carcinoma (HCC) cells to disulfidptosis, which is dependent on the activation of NRF2-SLC7A11 pathway. GH significantly increased the levels of NRF2 and promoted the transcription of NRF2 target gene, SLC7A11, through autophagy-mediated non-canonical mechanism. Under the condition of glucose starvation, GH-induced upregulation of SLC7A11 aggravated uptake of cysteine, disturbance of GSH synthesis, depletion of NADPH, and accumulation of disulfide molecules, ultimately leading to the formation of disulfide bonds between different cytoskeleton proteins and disulfidptosis eventually. Collectively, the findings underscore the potential role of GH in promoting cancer cell disulfidptosis, thereby offering a promising avenue for the treatment of drug-resistant HCC in clinical settings.
Collapse
Affiliation(s)
- Mengjiao Shi
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Xinyan Li
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ying Guo
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yinggang Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jiayi Xu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Liangwen Yan
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Rongrong Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Hong Wang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Shenkang Tang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Department of Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Yaping Zhao
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Zongfang Li
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yetong Feng
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Dongmei Ren
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Pengfei Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Key Laboratory of Environment and Genes Related To Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, 710061, China
| |
Collapse
|
17
|
Shimura T, Yin C, Ma R, Zhang A, Nagai Y, Shiratori A, Ozaki H, Yamashita S, Higashi K, Sato Y, Imaoka H, Kitajima T, Kawamura M, Koike Y, Okita Y, Yoshiyama S, Ohi M, Hayashi A, Imai H, Zhang X, Okugawa Y, Toiyama Y. The prognostic importance of the negative regulators of ferroptosis, GPX4 and HSPB1, in patients with colorectal cancer. Oncol Lett 2025; 29:144. [PMID: 39850719 PMCID: PMC11755263 DOI: 10.3892/ol.2025.14890] [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: 09/16/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
The prognostic value of negative regulators of ferroptosis in patients with colorectal cancer (CRC) has not yet been fully elucidated. The present study performed a systematic in silico identification and selection of candidate negative regulators of ferroptosis using The Cancer Genome Atlas data cohort (n=367), followed by clinical validation through immunohistochemistry of samples from patients with CRC (n=166) and further in vitro evaluation. In silico analysis identified specific light-chain subunit of the cystine/glutamate antiporter, AIFM2, NFE2L2, FTH1, GLS2, glutathione peroxidase 4 (GPX4) and heat shock protein β-1 (HSPB1) genes as possible candidates. Furthermore, patients with high expression of GPX4 or HSPB1 exhibited significantly worse overall survival (OS) compared with those with low expression (P<0.01 for both). Immunohistochemical analysis revealed that both OS and recurrence-free survival (RFS) of patients with CRC and high GPX4 or HSPB1 expression were significantly worse compared with in patients with low expression (P<0.01 for all). Furthermore, multivariate analysis showed that high GPX4 and HSPB1 expression were independent risk factors for poor oncological outcome for OS and RFS (GPX4: RFS, P=0.03; HSPB1: OS, P=0.006 and RFS, P<0.0001). Moreover, the effects of GPX4 and HSPB1 small interfering RNAs on two CRC cell lines (DLD-1 and SW480) indicated that GPX4 and HSPB1 may exhibit important roles in attenuating the cytotoxic effect of 5-fluorouracil-based chemotherapy. In conclusion, the current study confirmed that GPX4 and HSPB1 may serve as substantial prognostic- and recurrence-predictive biomarkers in patients with CRC.
Collapse
Affiliation(s)
- Tadanobu Shimura
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Chengzeng Yin
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Ruiya Ma
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Surgery, Tangshan Gongren Hospital, Tangshan, Hebei 063007, P.R. China
| | - Aiying Zhang
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yuka Nagai
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Aoi Shiratori
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hana Ozaki
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Shinji Yamashita
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Koki Higashi
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yuki Sato
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hiroki Imaoka
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Takahito Kitajima
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Genomic Medicine, Mie University Hospital, Tsu, Mie 514-8507, Japan
| | - Mikio Kawamura
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yuhki Koike
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yoshiki Okita
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Shigeyuki Yoshiyama
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masaki Ohi
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Akinobu Hayashi
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hiroshi Imai
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Xueming Zhang
- Department of Surgery, Tangshan Gongren Hospital, Tangshan, Hebei 063007, P.R. China
| | - Yoshinaga Okugawa
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Genomic Medicine, Mie University Hospital, Tsu, Mie 514-8507, Japan
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
18
|
Zhu F, Dan T, Hua S. KEAP1-NRF2/HO-1 Pathway Promotes Ferroptosis and Neuronal Injury in Schizophrenia. Brain Behav 2025; 15:e70311. [PMID: 40021790 PMCID: PMC11870791 DOI: 10.1002/brb3.70311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/31/2024] [Accepted: 01/16/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND This study investigates the role of the KEAP1-NRF2/HO-1 signaling pathway in inducing ferroptosis and contributing to neuronal damage in schizophrenia. METHODS We retrieved schizophrenia-related data and ferroptosis-related genes from the RNA microarray dataset GSE27383 and FerrDB database, respectively. Bioinformatics data identified KEAP1 as a downregulated gene, which was validated using qRT-PCR and Western blot. We assessed intracellular Fe2⁺ content, MDA levels, GSH, and GPX4 in the prefrontal cortex and peripheral blood mononuclear cells (PBMCs) of patients with schizophrenia. Cortical interneurons (cINs) were generated from human-induced pluripotent stem cells (hiPSCs) of patients with schizophrenia and used to explore KEAP1 alterations during neurodevelopment. In addition, KEAP1 overexpression was induced in cINs via transfection with pcDNA KEAP1. The intracellular Fe⁺ levels, oxidative stress indicators, lipid peroxidation, and inflammatory cytokines were measured after transfection. To investigate molecular mechanisms, KI696-a high-affinity probe that disrupts the KEAP1-NRF2 interaction-was applied, and changes in oxidative stress, lipid peroxidation (C11-BODIPY staining), iron metabolism, and inflammatory pathways were evaluated. RESULTS Patients with schizophrenia exhibited underexpression of KEAP1, a key regulator of ferroptosis, along with elevated intracellular Fe2⁺ levels and increased MDA concentrations, indicating enhanced lipid peroxidation and oxidative stress. Reduced GPX4 activity and GSH levels were also observed, suggesting an increased susceptibility to ferroptosis. To further explore this, cINs derived from hiPSCs of patients with schizophrenia were studied. These cells showed decreased KEAP1 expression. Overexpression of KEAP1 in cINs led to a reduction in intracellular Fe2⁺ concentrations and oxidative damage, highlighting KEAP1's regulatory role in ferroptosis. In addition, treatment with KI696 induced significant alterations in pathways related to oxidative stress, iron metabolism, antioxidant defenses, and inflammation. CONCLUSION Our findings indicate that the KEAP1-NRF2/HO-1 pathway contributes to ferroptosis and neuronal injury in schizophrenia.
Collapse
Affiliation(s)
- Feng Zhu
- Department of PsychiatryThe Second Affiliated Hospital of Hubei University of Science and TechnologyXianningHubeiChina
| | - Tangqun Dan
- Department of PsychiatryThe Second Affiliated Hospital of Hubei University of Science and TechnologyXianningHubeiChina
| | - Shuguang Hua
- Department of PsychiatryThe Second Affiliated Hospital of Hubei University of Science and TechnologyXianningHubeiChina
| |
Collapse
|
19
|
Wang Y, Zhang R, Wang A, Wang X, Wang X, Zhang J, Liu G, Huang K, Liu B, Hu Y, Pan S, Ruze X, Zhai Q, Xu Y. COPB1 deficiency triggers osteoporosis with elevated iron stores by inducing osteoblast ferroptosis. J Orthop Translat 2025; 51:312-328. [PMID: 40206560 PMCID: PMC11981772 DOI: 10.1016/j.jot.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/25/2024] [Accepted: 01/19/2025] [Indexed: 04/11/2025] Open
Abstract
Background Osteoporosis (OP) is a systemic bone metabolic disease that results from an imbalance between bone formation and bone resorption. The accumulation of iron has been identified as an independent risk factor for osteoporosis. Ferroptosis, a novel form of programmed cell death, is driven by iron-dependent lipid peroxidation. Nevertheless, the precise role of ferroptosis in iron accumulation-induced osteoporosis remains uncertain. Methods We utilized proteomics and ELISA to screen key regulatory molecules related to iron accumulation in osteoporosis populations. HE staining was used to assess osteocyte changes in Hamp knockout (KO) iron accumulation mouse models. Western Blot, qPCR, ALP staining, and Alizarin Red staining were employed to explore the effects of siRNA-mediated gene knockdown on osteogenic differentiation in the MC3T3 cell line. ELISA, micro-CT, von Kossa staining, toluidine blue staining, TRAP staining, and calcein analysis were used to study the bone phenotype of conditional gene knockout mice. RNA-seq, endoplasmic reticulum activity probes, transmission electron microscopy (TEM), Western Blot, co-immunoprecipitation (Co-IP), flow cytometry, and ChIP-seq were employed to investigate the regulatory mechanisms of the target gene in osteogenic differentiation. OVX and Hamp KO mice were used to establish osteoporosis models, and AAV-mediated overexpression was employed to explore the intervention effects of the target gene on osteoporosis. Results The experiments demonstrate that iron accumulation can lead to changes in COPB1 expression levels in bone tissue. Cellular and animal experiments revealed that COPB1 deficiency reduces the osteogenic ability of osteoblasts. Transcriptome analysis and phenotypic experiments revealed that COPB1 deficiency induces ferroptosis and endoplasmic reticulum stress in cells. Further investigation confirmed that COPB1 plays a key role in endoplasmic reticulum stress by inhibits SLC7A11 transcription via ATF6. This reduces cystine uptake, ultimately inducing ferroptosis. Overexpression of COPB1 can restore osteogenic function in both cells and mice. Conclusion This study elucidated the essential role of COPB1 in maintaining bone homeostasis and highlights it as a potential therapeutic target for treating iron accumulation-related osteoporosis. The translational potential of this article Our data elucidate the critical role of COPB1 in maintaining bone homeostasis and demonstrate that COPB1 can directly promote bone formation, making it a potential therapeutic target for the future treatment of osteoporosis.
Collapse
Affiliation(s)
- Yike Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ruizhi Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Aifei Wang
- Department of Orthopaedics, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu, China
| | - Xiao Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiongyi Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jiajun Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Gongwen Liu
- Department of Orthopaedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, China
| | - Kai Huang
- Orthopaedic Institute, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, China
| | - Baoshan Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yutong Hu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Sheng Pan
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xieyidai Ruze
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiaocheng Zhai
- Division of Spine Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Youjia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
20
|
Zou L, Zhang T, Yang C, Liu W, Fahira A, Yang D, Zheng B, Yao X, Liu Y, Huang Z. Downregulation of SLC7A11 by Bis(4-Hydroxy-3,5-Dimethylphenyl) Sulfone Induces Ferroptosis in Hepatocellular Carcinoma Cell. Mol Carcinog 2025; 64:580-596. [PMID: 39763283 DOI: 10.1002/mc.23874] [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: 05/11/2024] [Revised: 11/07/2024] [Accepted: 12/11/2024] [Indexed: 02/13/2025]
Abstract
The progression of tumors has been demonstrated to have a strong correlation with ferroptosis. Bis(4-hydroxy-3,5-dimethylphenyl) sulfone (TMBPS) has been shown to effectively inhibit the proliferation of hepatocellular carcinoma (HCC), but its underlying mechanism is not clear. In this study, ferrostatin-1 (Fer-1) was employed to explore whether the death of HCC cells caused by TMBPS is related to ferroptosis. The intracellular lipid peroxides, Fe2+, malondialdehyde (MDA), GSH/GSSG, mitochondrial morphology, and potential of HCC cells were detected after TMBPS treatment. The target of TMBPS was predicted by the molecular docking approach and verified via quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and cellular heat transfer assay (CETSA). Our results revealed that Fer-1 effectively reversed the cell death induced by TMBPS in HCC cells. Treatment with TMBPS induced typical ferroptosis features, including increased levels of intracellular lipid peroxides, Fe2+, and MDA, along with a decreased GSSH/GSH ratio and mitochondrial potential. These effects were reversed by overexpressing SLC7A11. These findings suggest that the cell death triggered by TMBPS in HCC cells is linked to ferroptosis, potentially mediated through the inhibition of SLC7A11 expression.
Collapse
Affiliation(s)
- Liyi Zou
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Dongguan, Guangdong, China
| | - Taomin Zhang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Dongguan, Guangdong, China
| | - Cui Yang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Dongguan, Guangdong, China
| | - Weijing Liu
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Dongguan, Guangdong, China
| | - Aamir Fahira
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Dongguan, Guangdong, China
| | - Dongli Yang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Dongguan, Guangdong, China
| | - Biao Zheng
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, China
| | - Xiaojun Yao
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Yi Liu
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Dongguan, Guangdong, China
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Dongguan, Guangdong, China
| |
Collapse
|
21
|
Ding Z, Li Z, Sun K, Liu Y, Fang Z, Sun S, Li C, Wang Z. Mitochondrial Regulation of Ferroptosis in Cancer Cells. Int J Biol Sci 2025; 21:2179-2200. [PMID: 40083691 PMCID: PMC11900798 DOI: 10.7150/ijbs.105446] [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: 10/18/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Ferroptosis is an iron-dependent nonapoptotic regulated cell death modality characterized by lethal levels of lipid peroxide accumulation and disrupted antioxidant systems. An increasing number of studies have revealed correlations between ferroptosis and the pathophysiology and treatment of cancer. Given the intricate involvement of mitochondria in ferroptosis, as suggested by previous studies, here, we review advances in understanding the roles of mitochondrial quality control and mitochondrial metabolism (including the roles of the TCA cycle, reactive oxygen species, iron metabolism, and lipid metabolism) in cancer-related ferroptosis and outline the molecular mechanism and clinical translation of mitochondria-related ferroptosis in cancer treatment. with the aim of promoting the precise utilization and prevention of ferroptosis in cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Shengrong Sun
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Chenyuan Li
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Zhong Wang
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
22
|
Yang G, Qian B, He L, Zhang C, Wang J, Qian X, Wang Y. Application prospects of ferroptosis in colorectal cancer. Cancer Cell Int 2025; 25:59. [PMID: 39984914 PMCID: PMC11846473 DOI: 10.1186/s12935-025-03641-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/07/2025] [Indexed: 02/23/2025] Open
Abstract
Colorectal cancer (CRC) is a serious threat to human health with the third morbidity and the second cancer-related mortality worldwide. It is urgent to explore more effective strategy for CRC because of the acquired treatment resistance from the non-surgical conventional therapies, including radiation, chemotherapy, targeted therapy and immunotherapy. Ferroptosis is a novel form of programmed cell death characterized by iron-dependent lipid peroxidation species (ROS) accumulation and has been identified as a promising target for cancer treatment, especially for those with treatment resistance. In this review, we mainly summarize the recent studies on the influence and regulation of ferroptosis by which (including gut microbiota) modulating the metabolism of iron, amino acid and lipid. Thus this analysis may provide potential targets for inducing CRC ferroptosis and shed lights on the future application of ferroptosis in CRC.
Collapse
Affiliation(s)
- Gen Yang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Boning Qian
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Liya He
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Chi Zhang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jianqiang Wang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xinlai Qian
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Henan Provincial Key Laboratory of Molecular Oncologic Pathology, Xinxiang, Henan, China.
| | - Yongxia Wang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Henan Provincial Key Laboratory of Molecular Oncologic Pathology, Xinxiang, Henan, China.
| |
Collapse
|
23
|
Zhu Y, Xu W, He Y, Yang W, Song S, Wen C. Therapeutic implications of endoplasmic reticulum stress gene CCL3 in cervical squamous cell carcinoma. Cell Biol Toxicol 2025; 41:47. [PMID: 39976849 PMCID: PMC11842515 DOI: 10.1007/s10565-024-09949-3] [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: 05/12/2024] [Accepted: 11/20/2024] [Indexed: 02/23/2025]
Abstract
This study investigated ERS-related gene expressions in CESC, identifying two molecular subtypes, P1 and P2, and constructing a precise prognostic model based on these subtypes. TCGA's whole-genome expression profiles were used to recognize these subtypes through the ConsensusClusterPlus method, further refining prognostic models with univariate and Lasso Cox regression analyses validated by the GSE39001 dataset. The study analyzed the expression distribution of ERS marker genes within T cell subgroups using scRNA-seq data (GSE168652), highlighting T cell diversity. The critical role of the CCL3 gene in prognostic models was examined explicitly in CD8 + T cells from healthy individuals and CESC patients. Elevated CCL3 levels were observed in patients' CD8 + T cells compared to healthy controls. Functional experiments involving CCL3 knockdown and overexpression in HeLa and SiHa CESC cell lines were conducted to investigate its impact on cell proliferation, migration, and invasion. These findings were subsequently validated in a nude mouse model. The results demonstrated that suppressing CCL3 inhibited cell proliferation, migration, and invasion significantly, while its overexpression promoted these processes. In the mouse model, CCL3 silencing reduced tumor growth and decreased Ki-67 labeling within the tumor tissues, indicating the therapeutic potential of targeting CCL3 in CESC treatment, possibly through CD8 + T cell regulation. This study contributes new prognostic assessment tools and personalized treatment options for CESC patients, paving the way for more targeted therapies in CESC by discovering the CCL3 gene, presenting significant clinical implications.
Collapse
Affiliation(s)
- Yingping Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Hangzhou, China
| | - Wei Xu
- College of Basic Medical Science, Zhejiang Chinese Medical University, 548 Binwen Rd, Hangzhou, 310053, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Hangzhou, China
| | - Yuanfang He
- College of Basic Medical Science, Zhejiang Chinese Medical University, 548 Binwen Rd, Hangzhou, 310053, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Hangzhou, China
| | - Wenjuan Yang
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Siyue Song
- College of Basic Medical Science, Zhejiang Chinese Medical University, 548 Binwen Rd, Hangzhou, 310053, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Hangzhou, China
| | - Chengping Wen
- College of Basic Medical Science, Zhejiang Chinese Medical University, 548 Binwen Rd, Hangzhou, 310053, China.
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
24
|
Liu Z, Liu Q, Zeng A, Song L. Regulatory function of endoplasmic reticulum stress in colorectal cancer: Mechanism, facts, and perspectives. Int Immunopharmacol 2025; 147:114024. [PMID: 39764998 DOI: 10.1016/j.intimp.2025.114024] [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: 10/08/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/29/2025]
Abstract
Colorectal cancer (CRC) is an exceedingly common and profoundly impactful malignancy of the digestive system, posing a grave threat to human health. Endoplasmic reticulum stress (ERS) is an intracellular biological reaction that mobilizes the unfolded protein response (UPR) to tackling dysregulation in protein homeostasis. This process subtly modulates the cell to either restore normal cellular function or steer it towards apoptosis. The high metabolic demands of CRC cells sculpt a rigorous tumor microenvironment (TME), compelling CRC cells to experience ERS. Adaptive responses induced by mild ERS furnish the necessary conditions for the survival of CRC cells, whereas the cell death mechanisms triggered by sustained ERS could be considered a prospective strategy for cancer therapy. Considering the complex regulation of ERS in cancer development, this article offers a comprehensive review of the molecular mechanisms through which ERS influences CRC fate. It provides crucial insights for exploring the role of ERS in the occurrence and progression of CRC, laying a new theoretical foundation for devising precise therapeutic strategies targeting ERS. Furthermore, by synthesizing extensive clinical and preclinical studies, we delve into therapeutic strategies targeting ERS, including the potential of targeting ERS in immunotherapy, the utilization of native compounds, advancements in proteasome inhibitors, and the potential synergies of these strategies with traditional chemotherapy agents and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Zihan Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan 610041, China.
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
25
|
Dastghaib S, Shafiee SM, Ramezani F, Ashtari N, Tabasi F, Saffari-Chaleshtori J, Siri M, Vakili O, Igder S, Zamani M, Niknam M, Nasery MM, Kokabi F, Wiechec E, Mostafavi-Pour Z, Mokarram P, Ghavami S. NRF-mediated autophagy and UPR: Exploring new avenues to overcome cancer chemo-resistance. Eur J Pharmacol 2025; 988:177210. [PMID: 39706466 DOI: 10.1016/j.ejphar.2024.177210] [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: 08/12/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
The development of chemo-resistance remains a significant hurdle in effective cancer therapy. NRF1 and NRF2, key regulators of redox homeostasis, play crucial roles in the cellular response to oxidative stress, with implications for both tumor growth and resistance to chemotherapy. This study delves into the dualistic role of NRF2, exploring its protective functions in normal cells and its paradoxical support of tumor survival and drug resistance in cancerous cells. We investigate the interplay between the PERK/NRF signaling pathway, ER stress, autophagy, and the unfolded protein response, offering a mechanistic perspective on how these processes contribute to chemoresistance. Our findings suggest that targeting NRF signaling pathways may offer new avenues for overcoming resistance to chemotherapeutic agents, highlighting the importance of a nuanced approach to redox regulation in cancer treatment. This research provides a molecular basis for the development of NRF-targeted therapies, potentially enhancing the efficacy of existing cancer treatments and offering hope for more effective management of resistant tumors.
Collapse
Affiliation(s)
- Sanaz Dastghaib
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, 7193635899, Shiraz, Iran
| | - Sayed Mohammad Shafiee
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, 7134845794, Shiraz, Iran
| | - Fatemeh Ramezani
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, 51664, Tabriz, Iran
| | - Niloufar Ashtari
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
| | - Farhad Tabasi
- Department of Neurosurgery, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Javad Saffari-Chaleshtori
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, 7134845794, Shiraz, Iran; Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, 8813833435, Shahrekord, Iran
| | - Morvarid Siri
- Autophagy Research Center, Shiraz University of Medical Sciences, 7134845794, Shiraz, Iran
| | - Omid Vakili
- Autophagy Research Center, Shiraz University of Medical Sciences, 7134845794, Shiraz, Iran; Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, 73461-81746, Isfahan, Iran
| | - Somayeh Igder
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, 6135715794, Ahvaz, Iran
| | - Mozhdeh Zamani
- Autophagy Research Center, Shiraz University of Medical Sciences, 7134845794, Shiraz, Iran
| | - Maryam Niknam
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, 7134845794, Shiraz, Iran
| | - Mahshid Moballegh Nasery
- Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), 7616911319, Tehran, Iran
| | - Fariba Kokabi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, 9177948564, Mashhad, Iran
| | - Emilia Wiechec
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555, Katowice, Poland; Department of Otorhinolaryngology in Linköping, Anaesthetics, Operations and Specialty Surgery Center, Region Östergotland, 58185, Linköping, Sweden
| | - Zohreh Mostafavi-Pour
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, 7134845794, Shiraz, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, 7134845794, Shiraz, Iran.
| | - Pooneh Mokarram
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, 7134845794, Shiraz, Iran.
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada; Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555, Katowice, Poland; Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB, R3E 0V9, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, R3E 0V9, Canada.
| |
Collapse
|
26
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2025; 480:759-784. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [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: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
27
|
Huang J, Zeng N, Xu S, Lv Y, Li X, Yang P, Liu Y. The study on bone marrow mesenchymal stem cell-derived extracellular matrix promoting the repair of damaged chondrocytes by regulating the Notch1/RBPJ pathway. Cytotechnology 2025; 77:35. [PMID: 39764424 PMCID: PMC11700074 DOI: 10.1007/s10616-024-00702-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/25/2024] [Indexed: 03/08/2025] Open
Abstract
Cartilage and joint damage can lead to cartilage degeneration. Bone marrow mesenchymal stem cells (BMSCs) have the potential to address cartilage damage. Hence, this study probed the mechanism of BMSC-extracellular matrix (BMSC-ECM) in promoting damaged chondrocyte repair by regulating the Notch1/RBPJ pathway. Human immortalized chondrocytes were cultured in vitro and treated with Notch1 small interfering (si)RNA, pCDNA3.1-Notch1, RBPJ siRNA and their negative controls (NCs). Damaged chondrocytes were constructed. Damaged chondrocyte-BMSC co-culture system was established and treated with lentiviral vector carrying short hairpin-Notch1 and its NC. Cell viability and apoptosis were assessed by CCK-8 and flow cytometry assays. Levels of glycosaminoglycan (GAG), Notch1 and RBPJ mRNA, and Notch1, RBPJ, Col2α1, mmp3, Hes1 and Hey1 were determined by a kit, RT-qPCR and Western blot. NICD nuclear translocation was detected by immunofluorescence. Damaged chondrocytes exhibited down-regulated Notch1 expression, reduced cell viability, and enhanced apoptosis. Further Notch1 knockdown aggravated chondrocyte damage, whereas its overexpression enhanced chondrocyte viability and decreased apoptosis. NICD translocated into the nucleus and bound to RBPJ to activate the Notch1 pathway. RBPJ silencing partly annulled Notch1-regulated damaged chondrocyte apoptosis. BMSC-damaged chondrocyte co-culture up-regulated Notch1, RBPJ, Col2α1, mmp3, Hes1, Hey1 and GAG levels, enhanced cell viability, and reduced apoptosis in chondrocytes, which were partly negated by Notch1 suppression, indicating that BMSC-ECM facilitated damaged chondrocyte repair by activating the Notch1/RBPJ pathway. BMSC-ECM promoted the repair of damaged chondrocytes by promoting NICD translocation into the nucleus and binding to RBPJ to activate the Notch1 pathway. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00702-6.
Collapse
Affiliation(s)
- Jiangfa Huang
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Ningjing Zeng
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, Guangdong China
| | - Shuchai Xu
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Yang Lv
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Xing Li
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Peng Yang
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Yan Liu
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| |
Collapse
|
28
|
Zhao Y, Zheng Y, Li H, Li Y, Wang R, Cai Y, Zheng H, Huo X, Ren J, Guo D, Luo R, Wu X, Lu J, Song Q, Zhang Y, Ma C, Wang L, Wang R, Wang J, He Y, Xu P, Sun J, Lu S. Protein folding dependence on selenoprotein M contributes to steady cartilage extracellular matrix repressing ferroptosis via PERK/ATF4/CHAC1 axis. Osteoarthritis Cartilage 2025; 33:261-275. [PMID: 39419437 DOI: 10.1016/j.joca.2024.10.005] [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: 05/31/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Initiation of endoplasmic reticulum (ER) stress is pivotal to the advancement of osteoarthritis (OA). We aimed to explore the function of ER-resident selenoprotein M (SELM) in cartilage-forming chondrocytes, investigating how SELM participates in cartilage extracellular matrix (ECM) metabolism and ER stress modulation. METHODS Articular cartilage samples with knee OA undergoing total knee arthroplasty were categorised into OA-smooth and OA-damaged groups, with primary chondrocytes extracted from smooth areas. Destabilization of the medial meniscus was induced in male C57BL6/J mice, with sham operations on the left knee as controls. After 8 weeks, knee joint tissues were collected for analysis. Histology and immunohistochemistry examined cartilage damage. Molecular biology techniques investigated how SELM affects ECM metabolism and ER stress regulation. RNA sequencing revealed the pathway changes after SELM intervention. AlphaFold demonstrated how SELM interacts with other molecules. Cultured cartilage explants helped determine the effects of SELM supplementation. RESULTS SELM expression was reduced in the damaged cartilage. Increasing SELM levels positively impacted ECM equilibrium. Decreasing SELM expression activated genes linked to degenerative ailments and impaired the cellular response to misfolded proteins, initiating the PERK/P-EIF2A/ATF4 pathway and exacerbating GSH/GSSG imbalance via the ATF4/CHAC1 axis. SELM likely participated in protein folding and modification by leveraging its thioredoxin domains. In vitro SELM supplementation mitigated IL-1β effects on damaged cartilage explants and suppressed beneficial chondrocyte phenotypes. CONCLUSIONS Our results confirm the involvement of SELM in ER stress-induced cartilage damage as well as protein folding, pointing to new directions in molecular therapy for degenerative diseases.
Collapse
Affiliation(s)
- Yitong Zhao
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Ying Zheng
- Department of Digestive Disease and Gastrointestinal Motility Research Room, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157 Xi Wu Road, Xi'an, Shaanxi 710004, PR China
| | - Han Li
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Yao Li
- Department of Pathology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, PR China
| | - Ru Wang
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Yongsong Cai
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, PR China
| | - Haishi Zheng
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, PR China
| | - Xinyu Huo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Jiajun Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Dongxian Guo
- Shaanxi Institute for Food and Drug Control, Xi'an, Shaanxi 710065, PR China
| | - Rui Luo
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Xinyao Wu
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Jingyi Lu
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Qingxin Song
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Yan Zhang
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Chenxing Ma
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Lu Wang
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Runyuan Wang
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| | - Jing Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Yingli He
- Department of Infectious Disease, The First Affiliated Teaching Hospital, SOM, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Peng Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, PR China
| | - Jian Sun
- Institute of Endemic Diseases, School of Public Health , Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, School of Public Health, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China.
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Street No.76, Xi'an, Shaanxi 710061, PR China
| |
Collapse
|
29
|
Safari MH, Rahimzadeh P, Alaei E, Alimohammadi M, Esfandiari N, Daneshi S, Malgard N, Farahani N, Taheriazam A, Hashemi M. Targeting ferroptosis in gastrointestinal tumors: Interplay of iron-dependent cell death and autophagy. Mol Cell Probes 2025; 79:102013. [PMID: 39837469 DOI: 10.1016/j.mcp.2025.102013] [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: 11/22/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
Ferroptosis is a regulated cell death mechanism distinct from apoptosis, autophagy, and necroptosis, marked by iron accumulation and lipid peroxidation. Since its identification in 2012, it has developed into a potential therapeutic target, especially concerning GI disorders like PC, HCC, GC, and CRC. This interest arises from the distinctive role of ferroptosis in the progression of diseases, presenting a new avenue for treatment where existing therapies fall short. Recent studies emphasize the promise of focusing on ferroptosis to fight GI cancers, showcasing its unique pathophysiological mechanisms compared to other types of cell death. By comprehending how ferroptosis aids in the onset and advancement of GI diseases, scientists aim to discover novel drug targets and treatment approaches. Investigating ferroptosis in gastrointestinal disorders reveals exciting possibilities for novel therapies, potentially revolutionizing cancer treatment and providing renewed hope for individuals affected by these tumors.
Collapse
Affiliation(s)
- Mohamad Hosein Safari
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Negin Esfandiari
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Neda Malgard
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
30
|
Zhou Y, Zeng L, Cai L, Zheng W, Liu X, Xiao Y, Jin X, Bai Y, Lai M, Li H, Jiang H, Hu S, Pan Y, Zhang J, Shao C. Cellular senescence-associated gene IFI16 promotes HMOX1-dependent evasion of ferroptosis and radioresistance in glioblastoma. Nat Commun 2025; 16:1212. [PMID: 39890789 PMCID: PMC11785807 DOI: 10.1038/s41467-025-56456-y] [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: 02/29/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025] Open
Abstract
Glioblastoma multiforme (GBM) remains a therapeutic challenge due to its aggressive nature and recurrence. This study establishes a radioresistant GBM cell model through repeated irradiation and observes a cellular senescence-like phenotype in these cells. Comprehensive genomic and transcriptomic analyses identify IFI16 as a central regulator of this phenotype and contributes to radioresistance. IFI16 activates HMOX1 transcription thereby attenuating ferroptosis by reducing lipid peroxidation, ROS production, and intracellular Fe2+ content following irradiation. Furthermore, IFI16 interacts with the transcription factors JUND and SP1 through its pyrin domain, robustly facilitating HMOX1 expression, further inhibiting ferroptosis and enhancing radioresistance in GBM. Notably, glyburide, a sulfonylurea compound, effectively disrupts IFI16 function and enhances ferroptosis and radiosensitivity. By targeting the pyrin domain of IFI16, glyburide emerges as a potential therapeutic agent against GBM radioresistance. These findings underscore the central role of IFI16 in GBM radioresistance and offer promising avenues to improve GBM treatment.
Collapse
Affiliation(s)
- Yuchuan Zhou
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Zeng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Linbo Cai
- Department of Neuro-Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Wang Zheng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinglong Liu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuqi Xiao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoya Jin
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yang Bai
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingyao Lai
- Department of Neuro-Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Hainan Li
- Department of Neuro-Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Hua Jiang
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, China
| | - Songling Hu
- Department of Preventive Dentistry, Shanghai Stomatological Hospital & School of Stomatology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
31
|
Wang C, You Z, Zhou G, Dong J, Tong S, Sun G. Amarogentin suppresses cell proliferation and EMT process through inducing ferroptosis in colorectal cancer. BMC Gastroenterol 2025; 25:46. [PMID: 39885392 PMCID: PMC11780999 DOI: 10.1186/s12876-025-03649-w] [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: 10/11/2024] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one common tumor with the high death rate, and badly affects the normal lives of CRC patients. Amarogentin (AG) has been found to exhibit regulatory roles and join into the progression of multiple diseases. However, the regulatory impacts and associated molecular mechanisms of AG in CRC progression keep unclear. METHODS AND RESULTS In this study, it was demonstrated that AG weakened CRC cell viability in a concentration- and time-dependent manner. In addition, AG accelerated cell apoptosis by triggering ferroptosis. The cell invasion and EMT process were restrained after AG treatment, but these impacts were reversed after Fer-1 addition. Moreover, it was uncovered that AG retarded Nrf2/HO-1/GPX4 activation. Additionally, AG modulated PTC cell viability and stimulated ferroptosis. At last, it was illustrated that AG suppressed tumor growth in vivo. CONCLUSION In conclusion, it was disclosed that AG suppressed cell proliferation and EMT process through inducing ferroptosis in CRC, and retarded Nrf2/HO-1/GPX4 activation. This discovery suggested that AG may be one effective drug for ameliorating CRC progression.
Collapse
Affiliation(s)
- Chao Wang
- Anhui Medical University, Hefei, 230022, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Zihao You
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Guoqing Zhou
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Juanjuan Dong
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Sihao Tong
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Guoping Sun
- Anhui Medical University, Hefei, 230022, China.
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
32
|
Yang X, Wu H, Liu D, Zhou G, Zhang D, Yang Q, Liu Y, Li Y. The link between ferroptosis and autophagy in myocardial ischemia/reperfusion injury: new directions for therapy. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10590-6. [PMID: 39885084 DOI: 10.1007/s12265-025-10590-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
Myocardial ischemia/reperfusion (I/R)-induced cell death, such as autophagy and ferroptosis, is a major contributor to cardiac injury. Regulating cell death may be key to mitigating myocardial ischemia/reperfusion injury (MI/RI). Autophagy is a crucial physiological process involving cellular self-digestion and compensation, responsible for degrading excess or malfunctioning long-lived proteins and organelles. During MI/RI, autophagy plays both "survival" and "death" roles. A growing body of research indicates that ferroptosis is a type of autophagy-dependent cell death. This article provides a comprehensive review of the functions of autophagy and ferroptosis in MI/RI, as well as the molecules mediating their interaction. Understanding the link between autophagy and ferroptosis may offer new therapeutic directions for MI/RI, bearing significant clinical implications.
Collapse
Affiliation(s)
- Xiaoting Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| | - Hui Wu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China.
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China.
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China.
| | - Di Liu
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huang Shi, HuBei Province, China
| | - Gang Zhou
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Dong Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Qingzhuo Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yanfang Liu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| | - Yi Li
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| |
Collapse
|
33
|
Dong X, Nie J, Huang A, Chen L, Zang E, Xiang Z, Hao X, Yan S, Ding X, Zhao Y. A novel small molecule NJH-13 induces pyroptosis via the Ca 2+ driven AKT-FOXO1-GSDME signaling pathway in NSCLC by targeting TRPV5. J Adv Res 2025:S2090-1232(25)00047-5. [PMID: 39832720 DOI: 10.1016/j.jare.2025.01.028] [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: 11/09/2024] [Revised: 01/05/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION Pyroptosis represents a mode of programmed necrotic cell death (PCD), mediated by members of gasdermin family (GSDMs), such as GSDME. It is emerging as a promising approach for combating cancer. Notably, GSDME is the key modulator for the switch between apoptosis and pyroptosis in cells. However, GSDME is often downregulated in many malignancies, including lung adenocarcinoma. OBJECTIVE To identify novel pyroptosis inducers in non-small cell lung cancer (NSCLC) and dissect the underlying mechanism. METHODS Pyroptosis was examined by live cell imaging, PI/Hoechst/Annexin V staining, LDH release assay, ELISA, and western blot assays. DARTS, CETSA, molecular docking was used to identify the target of NJH-13. RNA-seq, qPCR, chromatin immunoprecipitation (ChIP), dual luciferase assays were used elucidate the mechanism. RESULTS In this study, NJH-13, an N-containing heterocycle, was screened out and identified to possess the ability to activate GSDME, consequently triggering pyroptosis in NSCLC cells. By using the DARTS strategy, transient receptor potential cation channel subfamily V member 5 (TRPV5) was identified as a potential target of NJH-13. NJH-13 increased intracellular calcium level and triggered oxidative stress, both of which are critical events leading to pyroptosis mediated by GSDME. Mechanistically, NJH-13 enhanced the transcription of GSDME via the protein kinase B (AKT)/forkhead box transcription factor O1 (FOXO1) signaling pathway. ChIP revealed that FOXO1 bound directly to the promoter region of GSDME, thus triggering the GSDME-mediated pyroptosis. Pharmacological and genetic activation of AKT or inhibition of FOXO1 partially rescued NJH-13-induced pyroptotic cell death. Moreover, NJH-13 treatment suppressed tumor growth in vivo. CONCLUSION Taken together, our results revealed that TRPV5 is a distinctive target for manipulating pyroptosis and provided evidence that NJH-13 functions as a potential anti-cancer agent capable of triggering pyroptosis in NSCLC cells.
Collapse
Affiliation(s)
- Xianxiang Dong
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jiahui Nie
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Aiying Huang
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Li Chen
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Erkang Zang
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; School of Life Sciences, Yunnan University, Kunming 650091, PR China
| | - Zhengrui Xiang
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiaojiang Hao
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Shengjiao Yan
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China.
| | - Xiao Ding
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, PR China.
| | - Yuhan Zhao
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China.
| |
Collapse
|
34
|
Słoka J, Strzałka-Mrozik B, Kubica S, Nowak I, Kruszniewska-Rajs C. Influence of Mesalazine on Ferroptosis-Related Gene Expression in In Vitro Colorectal Cancer Culture. Biomedicines 2025; 13:219. [PMID: 39857803 PMCID: PMC11762154 DOI: 10.3390/biomedicines13010219] [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: 12/28/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Colorectal cancer (CRC) is one of the most common oncological disorders. Its fundamental treatments include surgery and chemotherapy, predominantly utilizing 5-fluorouracil (5-FU). Despite medical advances, CRC continues to present a high risk of recurrence, metastasis and low survival rates. Consequently, significant emphasis has been directed towards exploring novel types of cell death, particularly ferroptosis. Ferroptosis is characterized by iron imbalance and the accumulation of lipid peroxides and reactive oxygen species (ROS), leading to cellular damage and death. Thus, the discovery of safe inducers of ferroptosis, offering new hope in the struggle against CRC, remains crucial. In this study, we applied the concept of drug repositioning, selecting mesalazine (MES), a non-steroidal anti-inflammatory drug (NSAID), for investigation. Methods: The study was conducted on the colon cancer cell line DLD-1 and normal intestinal epithelial cells from the CCD 841 CoN cell line. Both cell lines were treated with MES solutions at concentrations of 10, 20, 30, 40, and 50 mM. Cytotoxicity was assessed using the MTT assay, while ferroptosis-related gene expression analysis was performed using oligonucleotide microarrays, with RT-qPCR used for validation. Results: MES effectively reduces the viability of DLD-1 cells while minimally affecting normal intestinal cells. Subsequent oligonucleotide microarray analysis revealed that MES significantly alters the expression of 56 genes associated with ferroptosis. Conclusions: Our results suggest that MES may induce ferroptosis in CRC, providing a foundation for further research in this area.
Collapse
Affiliation(s)
| | - Barbara Strzałka-Mrozik
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (J.S.); (S.K.); (I.N.); (C.K.-R.)
| | | | | | | |
Collapse
|
35
|
Xia W, Lv Y, Zou Y, Kang Z, Li Z, Tian J, Zhou H, Su W, Zhong J. The role of ferroptosis in colorectal cancer and its potential synergy with immunotherapy. Front Immunol 2025; 15:1526749. [PMID: 39850905 PMCID: PMC11754392 DOI: 10.3389/fimmu.2024.1526749] [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: 11/12/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and deadly malignancies worldwide. Recently, ferroptosis, a novel form of regulated cell death characterized by iron dependency and lipid peroxidation, has garnered significant attention from researchers. The mechanisms underlying ferroptosis, including intracellular iron levels, lipid peroxidation, and antioxidant system regulation, offer new insights into cancer treatment strategies. This study aims to explore the emerging role of ferroptosis in the context of immunotherapy for CRC, highlighting its potential mechanisms and clinical applications. We employed a comprehensive review of current literature to elucidate the biological mechanisms of ferroptosis, its relationship with CRC, and the interplay between ferroptosis and immunotherapy. Ferroptosis reshapes the tumor microenvironment (TME) by regulating intracellular iron levels, lipid metabolism, and antioxidant systems, significantly enhancing the efficacy of immune checkpoint inhibitors (ICIs). Meanwhile, traditional Chinese medicine therapies promote antitumor immunity by modulating the TME and inducing ferroptosis. Additionally, advances in nanotechnology have facilitated precise therapy by enabling targeted delivery of ferroptosis inducers or immunomodulators, transforming "cold" tumors into "hot" tumors and further boosting ICI efficacy. This study comprehensively reviews the latest developments in ferroptosis, immunotherapy, traditional Chinese medicine, and nanotechnology in CRC, highlighting the importance of ferroptosis-related biomarkers and novel inducers for personalized treatment. In summary, ferroptosis offers a promising strategy to overcome CRC therapy resistance and enhance immunotherapy efficacy, warranting further investigation and translational application.
Collapse
Affiliation(s)
- Wenhua Xia
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuanhao Lv
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yan Zou
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zhanting Kang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zhaoyi Li
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiaqi Tian
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Hongyan Zhou
- Xinxiang Key Laboratory of Precision Diagnosis and Treatment for Colorectal Cancer, Xinxiang First People’s Hospital, Xinxiang, China
| | - Wei Su
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Xinxiang Engineering Technology Research Center of Digestive Tumor Molecular Diagnosis, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Precision Diagnosis and Treatment for Colorectal Cancer, Xinxiang First People’s Hospital, Xinxiang, China
- Xinxiang Engineering Technology Research Center of Digestive Tumor Molecular Diagnosis, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
36
|
Pu X, Wu H, Liu X, Yang F. PRMT4 Reduced Erastin-Induced Ferroptosis in Nasopharyngeal Carcinoma Cisplatin-Resistant Cells by Nrf2/GPX4 Pathway. J Environ Pathol Toxicol Oncol 2025; 44:57-71. [PMID: 39462450 DOI: 10.1615/jenvironpatholtoxicoloncol.2024053754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is one of the common malignant tumors in clinic. In the current study, we aim to investigate the effects of PRMT4 on erastin-induced ferroptosis in NPC by cisplatin resistant. PRMT4 expression in patients with NPC by cisplatin was upregulated. PRMT4 upregulation promoted cell growth of erastin-induced ferroptosis in NPC cisplatin-resistant cells. PRMT4 downregulation reduced cell growth of erastin-induced ferroptosis in NPC cisplatin-resistant cells. PRMT4 promoted tumor volume in mice model of erastin-induced NPC by cisplatin. PRMT4 upregulation reduced erastin-induced ferroptosis in NPC cisplatin-resistant cells by mitochondrial damage. PRMT4 upregulation induced Nrf2 protein expression in model of erastin-induced NPC by cisplatin. Nrf2 reduced the effects of si-PRMT4 on cell growth of erastin-induced ferroptosis in NPC cisplatin-resistant cells. Nrf2 inhibitor reduced the effects of PRMT4 on cell growth of erastin-induced ferroptosis in NPC cisplatin-resistant cells. Nrf2 reduced the effects of si-PRMT4 on erastin-induced ferroptosis in NPC cisplatin-resistant cells by mitochondrial damage. PRMT4 protein interlinked with Nrf2 protein to decrease Nrf2 ubiquitination. Methylation increased PRMT4 DNA stability. Collectively, our data reveal that PRMT4 reduced erastin-induced ferroptosis in NPC cisplatin-resistant cells by Nrf2/GPX4 pathway, suggesting that targeting PRMT4 may present as a potential strategy against the development of NPC.
Collapse
Affiliation(s)
| | - Hong Wu
- Department of Otolaryngology, Xishan People's Hospital of Wuxi City, Wuxi 214105, China
| | - Xiaoyan Liu
- Department of Otolaryngology, Xishan People's Hospital of Wuxi City, Wuxi 214105, China
| | - Fang Yang
- Department of Otolaryngology, Xishan People's Hospital of Wuxi City, Wuxi 214105, China
| |
Collapse
|
37
|
Tian R, Guo S, Chen S, Wu J, Long A, Cheng R, Wang X, Huang L, Li C, Mao W, Xu P, Yu L, Pan H, Liu L. Natural products as Nrf2 modulators for ferroptosis inhibition in renal disease therapy: Recent progress and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156342. [PMID: 39742572 DOI: 10.1016/j.phymed.2024.156342] [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: 09/30/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2, NFE2L2) is a pivotal regulator of redox balance, metabolism, protein homeostasis and inflammation. Nrf2 is critically involved in both ferroptosis and renal diseases, and may serve as a significant target for many natural products in the treatment of renal diseases. However, a comprehensive overview on this topic is still lacking. PURPOSE To review the protective or therapeutic effects of natural products regulating Nrf2-related ferroptosis against various renal diseases. METHODS We systematically searched the electronic databases involving PubMed, Web of Science, Google Scholar, China National Knowledge Internet (CNKI), Wanfang Database and VIP Database. To ensure a comprehensive exploration, keywords including Nrf2, ferroptosis, natural products, phytochemicals, renal disease, kidney disease, kidney injury and nephropathy were employed. RESULTS Ferroptosis is deeply implicated in various kinds of renal diseases, notably including cisplatin-induced acute kidney injury, sepsis-associated acute kidney injury, renal ischemia/reperfusion injury, diabetic nephropathy, kidney stones and renal fibrosis. Nrf2 plays a regulatory role on many important genes related to iron metabolism, antioxidant system and lipid metabolism, thereby modulating ferroptosis. More than twenty natural products exert renoprotective effects by inhibiting ferroptosis via the regulation of Nrf2. This review presents a comprehensive overview of recent advancements in elucidating the ferroptosis involvement in renal diseases, the role of Nrf2 in regulating ferroptosis, and summarizes the renoprotective natural products as Nrf2 modulators for ferroptosis inhibition. CONCLUSION Through the comprehensive insights, this review clarifies the protective or therapeutic effects of natural products as Nrf2 modulators for ferroptosis inhibition in renal disease therapy, in the pursuit of providing new research ideas and directions for the treatment of renal diseases. Further drug development aimed at discovering more natural products and optimizing their utilization for disease treatment is necessary.
Collapse
Affiliation(s)
- Ruimin Tian
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Shan Guo
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shudong Chen
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiaqi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Aoyang Long
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ran Cheng
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiaowan Wang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Lihua Huang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Chuang Li
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Wei Mao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China
| | - Peng Xu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China
| | - Lili Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Hudan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China.
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China.
| |
Collapse
|
38
|
Wu J, Luo D, Tou L, Xu H, Jiang C, Wu D, Que H, Zheng J. NEK2 affects the ferroptosis sensitivity of gastric cancer cells by regulating the expression of HMOX1 through Keap1/Nrf2. Mol Cell Biochem 2025; 480:425-437. [PMID: 38503948 PMCID: PMC11695390 DOI: 10.1007/s11010-024-04960-y] [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: 11/17/2023] [Accepted: 02/05/2024] [Indexed: 03/21/2024]
Abstract
NEK2 is a serine/threonine protein kinase that is involved in regulating the progression of various tumors. Our previous studies have found that NEK2 is highly expressed in gastric cancer and suggests that patients have a worse prognosis. However, its role and mechanism in gastric cancer are only poorly studied. In this study, we established a model of ferroptosis induced by RSL3 or Erastin in AGS cells in vitro, and konckdown NEK2, HOMX1, Nrf2 by siRNA. The assay kit was used to analyzed cell viability, MDA levels, GSH and GSSG content, and FeRhoNox™-1 fluorescent probe, BODIPY™ 581/591 C11 lipid oxidation probe, CM-H2DCFDA fluorescent probe were used to detected intracellular Fe2+, lipid peroxidation, and ROS levels, respectively. Calcein-AM/PI staining was used to detect the ratio of live and dead cells, qRT-PCR and Western blot were used to identify the mRNA and protein levels of genes in cells, immunofluorescence staining was used to analyze the localization of Nrf2 in cells, RNA-seq was used to analyze changes in mRNA expression profile, and combined with the FerrDb database, ferroptosis-related molecules were screened to elucidate the impact of NEK2 on the sensitivity of gastric cancer cells to ferroptosis. We found that inhibition of NEK2 could enhance the sensitivity of gastric cancer cells to RSL3 and Erastin-induced ferroptosis, which was reflected in the combination of inhibition of NEK2 and ferroptosis induction compared with ferroptosis induction alone: cell viability and GSH level were further decreased, while the proportion of dead cells, Fe2+ level, ROS level, lipid oxidation level, MDA level, GSSG level and GSSG/GSH ratio were further increased. Mechanism studies have found that inhibiting NEK2 could promote the expression of HMOX1, a gene related to ferroptosis, and enhance the sensitivity of gastric cancer cells to ferroptosis by increasing HMOX1. Further mechanism studies have found that inhibiting NEK2 could promote the ubiquitination and proteasome degradation of Keap1, increase the level of Nrf2 in the nucleus, and thus promote the expression of HMOX1. This study confirmed that NEK2 can regulate HMOX1 expression through Keap1/Nrf2 signal, and then affect the sensitivity of gastric cancer cells to ferroptosis, enriching the role and mechanism of NEK2 in gastric cancer.
Collapse
Affiliation(s)
- Jianyong Wu
- Gastroenterology Department, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Desheng Luo
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Laizhen Tou
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Hongtao Xu
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Chuan Jiang
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Dan Wu
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Haifeng Que
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Jingjing Zheng
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China.
| |
Collapse
|
39
|
Wang X, Zhang T, Qu L, Zhang Y, Gao G. Auriculasin induces mitochondrial oxidative stress and drives ferroptosis by inhibiting PI3K/Akt pathway in non-small cell lung cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:967-977. [PMID: 39093464 DOI: 10.1007/s00210-024-03328-9] [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: 06/04/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Non-small cell lung cancer (NSCLC) accounts for the majority of cases of lung cancer with poor outcomes. Auriculasin is a prenylated isoflavone abundant in the root of F. philippinensis with multiple pharmacological effects, including anticancer role. However, its roles in NSCLC remain largely unknown. NSCLC A549 cells were treated with auriculasin in vitro, and used to induce xenograft models. Cell viability was detected via CCK-8 assay. Mitochondrial oxidative stress was analyzed by JC-1 staining, ROS staining, and levels of MDA, SOD and GSH. Ferroptosis was assessed via iron content, and levels of ACSL4, PTGS2, FSP1 and GPX4. The phosphorylation levels of PI3K and Akt were measured by western blot. Auriculasin reduced NSCLC cell viability. Auriculasin promoted mitochondrial oxidative stress by reducing mitochondrial membrane potential, SOD and GSH levels, and enhancing ROS and MDA contents. In addition, auriculasin induced ferroptosis via increasing iron, ACSL4 and PTGS3 levels, and decreasing FSP1 and GPX4 levels. Furthermore, the potential targets of auriculasin in NSCLC were enriched in PI3K/Akt signaling. Auriculasin blunted PI3K/Akt pathway activation by blocking the phosphorylation. Activated PI3K/Akt signaling by activator 740Y-P reversed the effects of auriculasin on mitochondrial oxidative stress and ferroptosis. Finally, auriculasin reduced NSCLC cell growth in xenograft models. Auriculasin facilitates mitochondrial oxidative stress and induces ferroptosis through inhibiting PI3K/Akt pathway in NSCLC.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, China
| | - Tao Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, China
| | - Lin Qu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, China
| | - Yifan Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, China
| | - Guizhou Gao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
40
|
Wu T, Ji M, Li T, Luo L. The molecular and metabolic landscape of ferroptosis in respiratory diseases: Pharmacological aspects. J Pharm Anal 2025; 15:101050. [PMID: 40034685 PMCID: PMC11873008 DOI: 10.1016/j.jpha.2024.101050] [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: 02/29/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 03/05/2025] Open
Abstract
Ferroptosis is a form of cell death that occurs when there is an excess of reactive oxygen species (ROS), lipid peroxidation, and iron accumulation. The precise regulation of metabolic pathways, including iron, lipid, and amino acid metabolism, is crucial for cell survival. This type of cell death, which is associated with oxidative stress, is controlled by a complex network of signaling molecules and pathways. It is also implicated in various respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), lung cancer, pulmonary fibrosis (PF), and the coronavirus disease 2019 (COVID-19). To combat drug resistance, it is important to identify appropriate biological markers and treatment targets, as well as intervene in respiratory disorders to either induce or prevent ferroptosis. The focus is on the role of ferroptosis in the development of respiratory diseases and the potential of targeting ferroptosis for prevention and treatment. The review also explores the interaction between immune cell ferroptosis and inflammatory mediators in respiratory diseases, aiming to provide more effective strategies for managing cellular ferroptosis and respiratory disorders.
Collapse
Affiliation(s)
- Tong Wu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Miaorong Ji
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
41
|
Lu CL, Liu J, Yang JF. LncRNA-XIST Promotes Lung Adenocarcinoma Growth and Inhibits Ferroptosis by Regulating GPX4. Mol Biotechnol 2025; 67:187-195. [PMID: 38153663 DOI: 10.1007/s12033-023-00993-8] [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: 06/11/2023] [Accepted: 11/13/2023] [Indexed: 12/29/2023]
Abstract
This study aimed to explore the regulatory effects and molecular mechanisms of long non-coding RNA X-inactive-specific transcript (LncRNA-XIST) in lung adenocarcinoma. si-XIST or glutathione peroxidase 4 (GPX4) plasmids were transfected in PC-9 cells to suppress LncRNA-XIST expression or over-express GPX4, respectively. The mRNA expression levels of LncRNA-XIST and GPX4 in lung adenocarcinoma tissues or cells were assessed using RT-qPCR. CCK-8 assay was performed to examine cell activity, and corresponding biochemical kits were used to measure the levels of Fe2+, reactive oxygen species (ROS), malondialdehyde (MDA) in cells. Western blot is used to examine relative protein expression of FANCD2, SLC7A11, and GPX4 in lung adenocarcinoma cells. The mRNA and protein expression levels of LncRNA-XIST in clinical tissues and cells of lung adenocarcinoma were significantly higher than those in adjacent tissues and normal cells. Functional analysis showed that knockdown of LncRNA-XIST notably weakened the viability of lung adenocarcinoma cells and promoted ferroptosis (manifested by significantly up-regulated levels of ROS, MDA, and Fe2+ and down-regulated the expression of SLC7A11 and FANCD2, P < 0.05). Further mechanism analysis revealed that knockdown of LncRNA-XIST markedly inhibited the expression of GPX4 in lung adenocarcinoma cells and that GPX4 was significantly over-expressed in clinical tissues and cells of lung adenocarcinoma. Notably, the expression of GPX4 was positively correlated with that of LncRNA-XIST. Over-expression of GPX4 remarkably promoted cell proliferation and inhibited ferroptosis in lung adenocarcinoma. Besides, the GPX4 over-expression reversed the LncRNA-XIST knockdown-induced ferroptosis and decrease in lung adenocarcinoma cell viability. LncRNA-XIST increases the activity of lung adenocarcinoma cells and inhibits ferroptosis by up-regulating GPX4. Knocking down LncRNA-XIST may be an effective treatment for lung adenocarcinoma.
Collapse
Affiliation(s)
- Chen-Lin Lu
- Department of Respiratory and Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Jie Liu
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Jun-Fa Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China.
| |
Collapse
|
42
|
Xu C, Wen S, Du X, Zou X, Leung ELH, Zhou G, Wu Q, Shen B. Targeting regulated cell death (RCD) with naturally derived sesquiterpene lactones in cancer therapy. Pharmacol Res 2025; 211:107553. [PMID: 39706282 DOI: 10.1016/j.phrs.2024.107553] [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: 09/30/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Regulated cell death (RCD) is a type of cell death modulated by specific signal transduction pathways. Currently, known RCD types include apoptosis, autophagy, ferroptosis, necroptosis, cuproptosis, pyroptosis, and NETosis. Mutations in cancer cells may prevent the RCD pathway; therefore, targeting RCD in tumors has become a promising therapeutic approach. Sesquiterpene lactones represent a diverse and extensive class of plant-derived phytochemicals that serve as potential sources for developing various drugs. Recent studies have shown that sesquiterpene lactones have promising potential in cancer treatment. This review systematically summarizes recent progress in the study of sesquiterpene lactones as antitumor agents, highlighting their role in targeting various RCD pathways, including those involved in apoptosis, autophagy, ferroptosis, necroptosis, and cuproptosis. The primary purpose of the present review is to provide a clear picture of the regulation of RCD by sesquiterpene lactones against different targets in various cancers, which will facilitate the development of new strategies for cancer therapy.
Collapse
Affiliation(s)
- Cong Xu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China; State Key Laboratory of Quality Research in Chinese Medicines and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao
| | - Shaodi Wen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China
| | - Xiaoyue Du
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China
| | - Xinhua Zou
- Department of Vascular and Tumor Interventional Medicine, Affiliated Hospital of Jining Medical University, Jining 272000, China
| | | | - Guoren Zhou
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao.
| | - Bo Shen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China; DongTai People's Hospital, Dongtai, Jiangsu, China.
| |
Collapse
|
43
|
Li Y, Liu C, Fang B, Chen X, Wang K, Xin H, Wang K, Yang SM. Ferroptosis, a therapeutic target for cardiovascular diseases, neurodegenerative diseases and cancer. J Transl Med 2024; 22:1137. [PMID: 39710702 DOI: 10.1186/s12967-024-05881-6] [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: 09/02/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The identification of ferroptosis represents a pivotal advancement in the field of cell death research, revealing an entirely novel mechanism of cellular demise and offering new insights into the initiation, progression, and therapeutic management of various diseases. Ferroptosis is predominantly induced by intracellular iron accumulation, lipid peroxidation, or impairments in the antioxidant defense system, culminating in membrane rupture and consequent cell death. Studies have associated ferroptosis with a wide range of diseases, and by enhancing our comprehension of its underlying mechanisms, we can formulate innovative therapeutic strategies, thereby providing renewed hope for patients.
Collapse
Affiliation(s)
- Yinghui Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Cuiyun Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bo Fang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xinzhe Chen
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Kai Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China.
| | - Kun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Su-Min Yang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
44
|
Tan W, Liang Z, Tan X, Tan G. Ginsenoside Rg1 improves cigarette smoke-induced ferroptosis in COPD by regulating PERK/ATF4 axis to inhibit endoplasmic reticulum stress. Biochem Biophys Res Commun 2024; 739:150946. [PMID: 39531905 DOI: 10.1016/j.bbrc.2024.150946] [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: 07/12/2024] [Revised: 09/26/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Ferroptosis plays a key role in the development of chronic obstructive pulmonary disease (COPD). Whether ginsenoside Rg1 improves cigarette smoke-induced COPD or whether ginsenoside Rg1 improves COPD by inhibiting ferroptosis remains unknown. METHODS BEAS-2B cells were exposed to cigarette solution (CSE) for 24 h and treated with ginsenoside Rg1, the ferroptosis inhibitor Fer-1, and the PERK inhibitor GSK. Cell viability, endoplasmic reticulum stress, mitochondrial morphology, membrane potential, reactive oxygen species (ROS), iron levels, and the expression of related proteins were detected using corresponding methods. A COPD mouse model was constructed using cigarette smoke (CS). Ginsenoside Rg1 and GSK were administered via tube feeding 15 days after successful modeling. Mouse lung tissues were evaluated by HE staining. The expression of inflammatory markers, ROS, iron content, and related proteins was detected using corresponding methods. RESULTS The results demonstrated that in the CSE-exposed BEAS-2B cell model and CS-induced mouse COPD model, the expression levels of endoplasmic reticulum stress (ERS)-related factors such as GRP78 were increased, while those of the antioxidant markers GPX4 and GSH were significantly decreased. Ginsenoside Rg1 improved emphysema and inflammation by inhibiting ferroptosis in vivo and in vitro. Using a PERK inhibitor, we found that ginsenoside Rg1 inhibited ferroptosis in vivo and in vitro by regulating ERS. CONCLUSION This study showed that ginsenoside Rg1 alleviates cigarette smoke-induced COPD by regulating the PERK/ATF4 axis to inhibit ERS and ferroptosis.
Collapse
Affiliation(s)
- Wei Tan
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Zicheng Liang
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaoning Tan
- Department of Oncology, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China.
| | - Guangbo Tan
- Department of Pulmonology, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China.
| |
Collapse
|
45
|
Sui X, Gao B, Zhang L, Wang Y, Ma J, Wu X, Zhou C, Liu M, Zhang L. Scutellaria barbata D.Don and Hedyotis diffusa Willd herb pair combined with cisplatin synergistically inhibits ovarian cancer progression through modulating oxidative stress via NRF2-FTH1 autophagic degradation pathway. J Ovarian Res 2024; 17:246. [PMID: 39702302 DOI: 10.1186/s13048-024-01570-6] [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: 06/19/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Cisplatin (DDP) is one of the most effective anticancer drugs, commonly used to treat advanced ovarian cancer (OC). However, DDP has significant limitations of platinum-based drugs, including chemical resistance and high-dose toxic side effects. Traditional Chinese medicines (TCMs) often presented in the form of formula, in which the herb pair was the basic unit. Scutellaria barbata D.Don and Hedyotis diffusa Willd (SB-HD) are famous TCMs herb pair that have been shown to help treat multiple types of cancers. However, the synergistic effects and mechanism of combination of SB-HD and DDP to enhance DDP chemosensitivity in OC are still unknown. RESULTS In vitro, we found that the optimal proportion of SB-HD to inhibit the proliferation of OC cells was 2:1, SB-HD and DDP were shown to synergistically reduce the viability of OC cells, inhibit the colony formation, promote cell cycle arrest and apoptosis, as well as inhibit cell migration and invasion. In vivo, combination treatment significantly inhibited the growth of subcutaneous tumors in BALB/c nude mice and reduced the toxic side effects of DDP. Mechanistically, SB-HD and DDP combination treatment significantly promoted oxidative stress response, decreased MMP, inhibited ATP production, decreased ROS levels and increased SOD activity, increased the expression of NRF2, HO-1, ATG5 and LC3, decreased the expression of p62 and FTH1 both in OC cells and tumor tissue of mice. Inhibitor 3-MA (Methyladenine, autophagy inhibitor) and Fer-1 (Ferrostatin-1, iron ion inhibitor) can effectively reverse the expression changes of the key target proteins, but not ZnPP (Zinc protoporphyrin, HO-1 inhibitor). Through bioinformatics analysis, it was found that the abnormal expression level of NRF2 and FTH1 mRNA has a high prognostic value, at the same time, the other four key proteins respectively or interacting with NRF2 and FTH1, also play important roles in the occurrence and development of OC. CONCLUSION Our findings uncover a synergistic effect of SB-HD and DDP against OC through modulating oxidative stress via NRF2-FTH1 autophagic degradation pathway, which may provide an important theoretical foundation for the use of SB-HD and a new strategy for enhancing DDP chemosensitivity as well as reducing toxic side effects.
Collapse
Affiliation(s)
- Xue Sui
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Bingqing Gao
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- School of Pharmacy, Anhui Xinhua University, Hefei, 230088, China
| | - Liu Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- Department of Dermatology, Dalian Lvshunkou District Hospital of Traditional Chinese Medicine, Dalian, 116041, China
| | - Yanmin Wang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Junnan Ma
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xingchen Wu
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Chenyu Zhou
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Min Liu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| | - Lin Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
46
|
Kong L, Zhao M, Zhu X, Liu J, Zhang D, Ye Y. A Novel ⋅OH-Monitor ER-Targeted Probe to Expose the Function of Sorafenib. Chem Asian J 2024; 19:e202400980. [PMID: 39316060 DOI: 10.1002/asia.202400980] [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: 08/11/2024] [Revised: 09/21/2024] [Accepted: 09/22/2024] [Indexed: 09/25/2024]
Abstract
The hydroxyl radical (⋅OH), widely recognized as the most potent free radical, plays a crucial role in numerous physiological and pathological pathways due to its strong oxidizability.Ferroptosis, as a novel mode of cell death, is initiated by the accumulation of iron-dependent lipid peroxidation. Among them, ⋅OH as the original reactive oxygen species (ROSs)is mass-produced due to Fenton reaction in vivo and closely related to cancer treatment.Besides, endoplasmic reticulum (ER) as a membrane-rich structure organelle, is a crucial organelle in all eukaryotes where excessive expression of ROSs, including ⋅OH can triggerER stress which was reported thatwasclosely related toferroptosis. So developing a new probe for their interrelationship research is important. In this paper, we constructed a1,8-naphthalimide-based ER-targeted fluorescence probe named M-1 to monitor ⋅OH variation in vitro and vivo. What's more, we achieved the monitor of ⋅OH during ER stress andferroptosis processesin cancer cells, andfurther explored the important role of ER stress and ferroptosis processes in SF (sorafenib) involved cancer cells.
Collapse
Affiliation(s)
- Lingyu Kong
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Manfen Zhao
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaofei Zhu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| | - Jianfei Liu
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Di Zhang
- Henan Key Laboratory of Grain Quality and Safety Testing, Institute of Quality and Safety for Agro-products, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Yong Ye
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
47
|
Wang H, Su Z, Qian Y, Shi B, Li H, An W, Xiao Y, Qiu C, Guo Z, Zhong J, Wu X, Chen J, Wang Y, Zeng W, Zhan L, Wang J. Pentraxin-3 modulates hepatocyte ferroptosis and the innate immune response in LPS-induced liver injury. MOLECULAR BIOMEDICINE 2024; 5:68. [PMID: 39666228 PMCID: PMC11638432 DOI: 10.1186/s43556-024-00227-6] [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: 06/25/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
The liver plays a crucial role in the immune response during endotoxemia and is one of the critical targets for sepsis-related injuries. As a secretory factor involved in inflammation, pentraxin-3 (PTX3) has been demonstrated to regulate hepatic homeostasis; however, the relationship between PTX3 and cell crosstalk between immune cells and hepatocytes in the liver remains incompletely understood. In this study, we revealed that, compared with WT mice, Ptx3-/- mice with lipopolysaccharide (LPS)-induced endotoxemia exhibited alleviated liver damage, with reduced serum alanine transaminase and aspartate transaminase levels and an improved survival rate. Mechanistically, RNA-Seq and western blot results revealed that Ptx3 knockdown in hepatocytes increased the expression of Tfrc and Ccl20; consequently, Ptx3 deficiency regulated LPS-induced hepatocyte ferroptosis via increased mitochondrial reactive oxygen species and Fe2+ and recruited more macrophages by CCL20/CCR6 axis to be involved in inflammation and the clearance of harmful substances. Moreover, western blot and immunofluorescence staining confirmed that the NF-κB signaling pathway was upregulated upon LPS treatment in Ptx3-knockdown macrophages, promoting phagocytosis and polarization toward M1 macrophages. Collectively, our findings show that the absence of Ptx3 can ameliorate sepsis-induced liver injury by regulating hepatocyte ferroptosis and promote the recruitment and polarization of M1 macrophages. These findings offer a key basis for the development of effective treatments for acute infections.
Collapse
Affiliation(s)
- Huitong Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhaojie Su
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yunyun Qian
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Baojie Shi
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hao Li
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wenbin An
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yi Xiao
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Cheng Qiu
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhixiang Guo
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jianfa Zhong
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xia Wu
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jiajia Chen
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ying Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wei Zeng
- Department of Gastroenterology, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Linghui Zhan
- Department of Critical Care Medicine, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, 361004, China.
| | - Jie Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
48
|
Yue X, Pang M, Chen Y, Cheng Z, Zhou R, Wang Y, Zha Z, Huang L. Puerarin alleviates symptoms of preeclampsia through the repression of trophoblast ferroptosis via the CREB/HO-1 pathway. Placenta 2024; 158:145-155. [PMID: 39490110 DOI: 10.1016/j.placenta.2024.10.013] [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: 07/01/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Preeclampsia (PE) is a pregnancy-associated complication characterised by new-onset hypertension and proteinuria. This study explored the therapeutic potential of puerarin (Pue) in PE and investigated the underlying mechanism, with a focus on placental ferroptosis. METHODS Using an NG-nitro-L-arginine methyl ester (L-NAME)-induced PE mouse model, we assessed the effects of Pue on PE phenotypes and placental ferroptosis. Antioxidative and anti-ferroptotic effects of Pue were studied in three ferroptotic cell models (hypoxia/reperfusion, cobalt chloride, and erastin). The regulation of Pue on cAMP response element binding protein (CREB) and heme oxygenase-1 (HO-1) was evaluated through gain- and loss-of-function assays. Luciferase assays were used to elucidate the effect of Flag-CREB on Hmox1 promoter fragments. CREB/HO-1 modulation by Pue was validated in mouse placentas with PE. RESULTS Pue significantly alleviated maternal hypertension, proteinuria, fetal growth restriction, and placental damage in PE mice. This was associated with an upregulation of the anti-ferroptosis system (glutathione peroxidase 4 [GPX4], cys2/glutamate antiporter [SLC7A11], and glutathione [GSH]) and repression of reactive oxygen species (ROS) and malondialdehyde (MDA) in trophoblasts. Pue reduced HO-1 and CREB, and HO-1 deficiency upregulated GPX4 and SLC7A11. Manipulation of CREB expression led to changes in HO-1/GPX4; whereas, the regulation reversed by Pue administration. Flag-CREB enhanced luciferase activity on the full length Hmox1 promoter (-2000/+78), which contains three CREB1 binding sites (S1-S3). In contrast, no increase in luciferase activity was observed with promoter fragments (-850/+78) and (-550/+78), which contain only the CREB1 binding sites S2 and S3, respectively. DISCUSSION Pue ameliorated PE-like symptoms in mice by repressing trophoblast ferroptosis via inhibition of CREB signalling and affecting the Homx1 promoter.
Collapse
Affiliation(s)
- Xiaojing Yue
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Menglan Pang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yun Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhixing Cheng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ruisi Zhou
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhiqiang Zha
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
49
|
Zhang R, Zhang X, Zhu X, Li T, Li Y, Zhang P, Chen Y, Li G, Han X. Nanoparticles transfected with plasmid-encoded lncRNA-OIP5-AS1 inhibit renal ischemia-reperfusion injury in mice via the miR-410-3p/Nrf2 axis. Ren Fail 2024; 46:2319327. [PMID: 38419565 PMCID: PMC10906121 DOI: 10.1080/0886022x.2024.2319327] [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: 08/17/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024] Open
Abstract
Nanostructures composed of liposomes and polydopamine (PDA) have demonstrated efficacy as carriers for delivering plasmids, effectively alleviating renal cell carcinoma. However, their role in acute kidney injury (AKI) remains unclear. This study aimed to investigate the effects of the plasmid-encoded lncRNA-OIP5-AS1@PDA nanoparticles (POP-NPs) on renal ischemia/reperfusion (RI/R) injury and explore the underlying mechanisms. RI/R or OGD/R models were established in mice and HK-2 cells, respectively. In vivo, vector or POP-NPs were administered (10 nmol, IV) 48 h after RI/R treatment. In the RI/R mouse model, the OIP5-AS1 and Nrf2/HO-1 expressions were down-regulated, while miR-410-3p expression was upregulated. POP-NPs treatment effectively reversed RI/R-induced renal tissue injury, restoring altered levels of blood urea nitrogen, creatinine, malondialdehyde, inflammatory factors (IL-8, IL-6, TNF-α), ROS, apoptosis, miR-410-3p, as well as the suppressed expression of SOD and Nrf2/HO-1 in the model mice. Similar results were obtained in cell models treated with POP-NPs. Additionally, miR-410-3p mimics could reverse the effects of POP-NPs on cellular models, partially counteracted by Nrf2 agonists. The binding relationship between OIP5-AS1 and miR-410-3p, alongside miR-410-3p and Nrf2, has been substantiated by dual-luciferase reporter and RNA pull-down assays. The study revealed that POP-NPs can attenuate RI/R-induced injury through miR-410-3p/Nrf2 axis. These findings lay the groundwork for future targeted therapeutic approaches utilizing nanoparticles for RI/R-induced AKI.
Collapse
Affiliation(s)
- Rongjie Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Xin Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Xuhui Zhu
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Tao Li
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Yansheng Li
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Peng Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Yuanhao Chen
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Gao Li
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Xiuwu Han
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| |
Collapse
|
50
|
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
|