1
|
Yang J, Liu Y, He L, Yu W, Liu H, Chen T. X-ray-Sensitive Selenium Nanoparticles Enhance Esophageal Squamous Cell Carcinoma Radiotherapy through Activating P53/IGFBP3 Pathway by Regulating GPX2. ACS APPLIED MATERIALS & INTERFACES 2025; 17:24865-24876. [PMID: 40241246 DOI: 10.1021/acsami.4c22183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Radiotherapy remains a crucial treatment for esophageal squamous cell carcinoma (ESCC), although the development of radiation resistance and the occurrence of radiation-induced side effects pose significant clinical challenges. Selenium (Se) has obvious antitumor effects, but the sensitizing effect and mechanism of Se nanoparticles in ESCC radiotherapy remain to be determined. The aim of this study was to investigate which form of Se have superior sensitization of ESCC and to investigate how Se nanoparticles (LNT-SeNPs) can enhance the radiosensitivity of ESCC. Our findings indicate that LNT-SeNPs exhibit remarkable radiosensitizing activity with a higher safety index. These nanoparticles effectively inhibit cell growth, induce S-phase arrest, and promote apoptosis through increased reactive oxygen species (ROS) production. Furthermore, analysis via the GEO database revealed the correlation between the selenoprotein GPX2 and the radiosensitivity of esophageal cancer. Further investigations demonstrate that LNT-SeNPs suppress GPX2 expression, leading to apoptosis in ESCC cells via the p53/IGFBP3 signaling pathway. In conclusion, this study elucidates that LNT-SeNPs can enhance the effectiveness of radiotherapy for esophageal cancer, providing valuable insights into the potential use of Se-based drugs as adjunctive therapy. These findings pave the way for future clinical applications aimed at improving therapeutic outcomes in patients undergoing radiotherapy for ESCC.
Collapse
Affiliation(s)
- Jianwei Yang
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Ying Liu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- Department of Neurology, The First Affiliated Hospital of Jinan University, Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Lizhen He
- Department of Neurology, The First Affiliated Hospital of Jinan University, Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Wenfang Yu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- Department of Neurology, The First Affiliated Hospital of Jinan University, Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Hongmei Liu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Tianfeng Chen
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- Department of Neurology, The First Affiliated Hospital of Jinan University, Department of Chemistry, Jinan University, Guangzhou 510632, China
| |
Collapse
|
2
|
Tan Y, Yang L, Xu T, Wang Q, Huang M, Zhao Z, Chen X, Tang C, Tan W. GPX2 inhibition enhances antitumor efficacy of lenvatinib via promoting immunogenic cell death in hepatocellular carcinoma. J Transl Med 2025; 23:456. [PMID: 40251668 PMCID: PMC12007242 DOI: 10.1186/s12967-025-06468-5] [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: 02/13/2025] [Accepted: 04/06/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Immunogenic cell death (ICD) is a distinct subtype of regulatory cell death, and represents a potential mechanism to remodel the tumor microenvironment. Lenvatinib is established as the first-line therapy for advanced hepatocellular carcinoma (HCC), but drug resistance limits its efficiency. Our previous research showed that lenvatinib can inhibit GPX2 expression and induce reactive oxygen species (ROS)-related cell apoptosis in HCC. The present study intends to explore whether lenvatinib can induce ICD and clarify its underlying mechanisms in HCC. METHODS Flow cytometry was utilized to detect the expression levels of CRT and CD markers, measure intracellular ROS levels, and assess cell apoptosis. Western blot analysis was employed to determine changes in protein levels, while qRT-PCR analysis was used to quantify alterations in mRNA levels. Subcutaneous allograft tumor models were established to investigate the mechanism of lenvatinib against HCC. Immunohistochemical (IF) staining were used to detect the ratio of CD8+GZMB+ cells. RESULTS Herein, we found that HCC cells treated with lenvatinib or si-GPX2 showed increased ICD markers, such as CRT exposure, ATP secretion, and HMGB1 release. Notably, we demonstrated that lenvatinib promoted dendritic cells (DCs) maturation and CD8+ T cells activation, thus inducing HCC cell apoptosis when co-cultured with peripheral blood mononuclear cells. Additionally, we further demonstrated that lenvatinib or GPX2 inhibition triggers endoplasmic reticulum stress (ERS) in HCC cells, which is mediated by the accumulation of ROS. Our findings indicate that pre-treatment with the antioxidant N-acetylcysteine suppressed lenvatinib-induced expression of CRT on the cell membrane, ATP secretion and HMGB1 release, and inhibited lenvatinib-induced cell apoptosis. Furthermore, we also found that ERS inhibitor ISRIB could reverse lenvatinib-induced upregulation of ICD biomarkers. Moreover, we further identified that downregulation of GPX2 enhanced the efficacy of lenvatinib via triggering ERS-mediated ICD in HCC. CONCLUSIONS This study uncovered that lenvatinib could be a potent ICD inducer, which could trigger ERS via increasing ROS levels in HCC cells, which present valuable insights into the mechanism of lenvatinib-induced ICD in HCC cells. Collectively, our findings highlight the significant therapeutic potential of the combination of targeting GPX2 and treatment with lenvatinib for HCC.
Collapse
Affiliation(s)
- Yingzheng Tan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Lei Yang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui, China
| | - Tao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Medical Center of Digestive Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Qingbin Wang
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Meiyuan Huang
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Zhijian Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Center of Digestive Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Xun Chen
- Department of Hepatobiliary and Pancreatic Surgery, Medical Center of Digestive Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China.
| | - Caixi Tang
- Department of Hepatobiliary and Pancreatic Surgery, Medical Center of Digestive Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China.
| | - Wenliang Tan
- Department of Hepatobiliary and Pancreatic Surgery, Medical Center of Digestive Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China.
| |
Collapse
|
3
|
Zhang W, Hong X, Xiao Y, Wang H, Zeng X. Sorafenib resistance and therapeutic strategies in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2025; 1880:189310. [PMID: 40187502 DOI: 10.1016/j.bbcan.2025.189310] [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: 11/08/2024] [Revised: 03/30/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent and lethal cancers globally. While surgical resection and liver transplantation offer potential cures for early-stage HCC, the majority of patients are diagnosed at advanced stages where such interventions are not viable. Sorafenib, a multi-target kinase inhibitor, has been a cornerstone in the treatment of advanced HCC since its approval in 2007. Despite its significant clinical impact, less than half of the treated patients derive long-term benefits due to the emergence of resistance and associated side effects. This review focuses on the role of sorafenib, an FDA-approved multi-target kinase inhibitor, in treating advanced HCC, discusses the mechanisms underlying its therapeutic effects and associated resistance, and explores additional therapeutic strategies being investigated to improve patient outcomes.
Collapse
Affiliation(s)
- Weijing Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Xuechuan Hong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuling Xiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaodong Zeng
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China.
| |
Collapse
|
4
|
Xuan F, Zhao X, Pang W, Li Z, Yin X, Xie W, Zeng X, Nie L, Yang J, Li S, Lai P, Fang C. Biomimetic Co-delivery of Lenvatinib and FePt Nanoparticles for Enhanced Ferroptosis/Apoptosis Treatment of Hepatocellular Carcinoma. Adv Healthc Mater 2025; 14:e2401747. [PMID: 40114524 PMCID: PMC12023810 DOI: 10.1002/adhm.202401747] [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: 02/16/2025] [Indexed: 03/22/2025]
Abstract
Lenvatinib, endorse as a first-line targeted therapy, has demonstrated efficacy in extending the survival span of individuals afflicted with advanced Hepatocellular carcinoma (HCC). However, its therapeutic effect wears off with time, which is ascribed to the cancer cell's tendency to evade and tamper with its usual modes of action, severely limiting its clinical use. This study devises an innovative therapeutic modality involving the synergistic co-delivery of FePt nanoparticles (NPs) and Lenvatinib via poly lactic-co-glycolic acid (PLGA) NPs encase in HCC cell membranes (Len/FePt@CMP NPs). The investigation explores the mechanism through which Lenvatinib induces ferroptosis in HCC, notably by dampening the glutathione peroxidase 4 (GPX4) through the inhibition of fibroblast growth factor receptor 4. FePt NPs are engineered to enhance the efficacy of ferroptosis and apoptosis for HCC treatment. Concurrently, the incorporation of the cancer cell membrane facilitates the targeted accumulation of NPs at the tumor site, leveraging mechanisms of immune evasion and homologous targeting. This enhances ferroptosis/apoptosis treatment efficacy, triggeres by Len/FePt@CMP NPs, is convincingly demonstrated both in vitro and in vivo. The proposed approach has the potential to redefine HCC therapeutic paradigms by overcoming mono-therapeutic limitations in current clinical treatments, showcasing the improved efficacy of a comprehensive strategy.
Collapse
Affiliation(s)
- Feichao Xuan
- Department of Hepatobiliary Surgery IZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Xingyang Zhao
- Department of Hepatobiliary Surgery IZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Weiran Pang
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Zirong Li
- Department of Hepatobiliary Surgery IZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Xiangyi Yin
- Department of Hepatobiliary Surgery IZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Weizhong Xie
- Department of Hepatobiliary Surgery IZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Xiaojun Zeng
- Department of Hepatobiliary Surgery IZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Liming Nie
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Junying Yang
- Department of Hepatobiliary Surgery IZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Shiying Li
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Puxiang Lai
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Chihua Fang
- Department of Hepatobiliary Surgery IZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| |
Collapse
|
5
|
Yi N, Zhang L, Huang X, Ma J, Gao J. Lenvatinib-activated NDUFA4L2/IL33/PADI4 pathway induces neutrophil extracellular traps that inhibit cuproptosis in hepatocellular carcinoma. Cell Oncol (Dordr) 2025; 48:487-504. [PMID: 39585643 PMCID: PMC11996955 DOI: 10.1007/s13402-024-01013-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Lenvatinib is a potent first-line therapy for patients with hepatocellular carcinoma (HCC), but it also increased the number of neutrophils in HCC tumor microenvironment. METHODS CitH3, MPO-DNA, elastase and MPO activity were measured for assessing neutrophil extracellular traps (NETs) in vivo and in vitro. Cell cuproptosis was assessed by measurement of copper content, FDX1, and pyruvate. The functions of lenvatinib, DNase I, interleukin 33 (IL33) neutralizing antibody and GPX4 in tumor growth were explored in mice. RESULTS Lenvatinib induced NETs in the HCC tumor microenvironment via HCC cells, but not through the direct stimulation of neutrophils. In addition, NET clearance by DNase I improves the efficacy of lenvatinib therapy in HCC mouse models. Mechanistically, lenvatinib promoted the expression and secretion of IL33 by HCC cells that triggered NET formation. Moreover, IL33 knockdown in Hepa1-6 cells improved lenvatinib efficacy in Hepa1-6-bearing HCC model mice and reduced NET formation in the tumor microenvironment. Subsequently, lenvatinib increased IL33 production by increasing the NDUFA4L2 expression in HCC cells. Furthermore, we found that IL33 triggered NET formation in neutrophils by increasing the protein expression of PADI4 via the Akt/mTOR signaling pathway. Rapamycin inhibition of mTOR reduced PADI4 expression and NET formation. Consistently, PADI4 inhibition by the selective PAD4 inhibitor GSK484 hydrochloride (GSK484) improved lenvatinib response to HCC therapy. Importantly, NETs contribute to lenvatinib resistance by inhibiting cuproptosis, but not apoptosis, pyroptosis, or ferroptosis in HCC cells. Treatment with GSK484 reversed the inhibitory effects of NETs on cuproptosis and sensitized the HCC cells to lenvatinib. CONCLUSIONS Our study revealed that lenvatinib-induced NETs inhibited the cuproptosis of HCC cells, suggesting that targeting the IL33/PADI4/NET axis represents a promising therapeutic strategy for ameliorating lenvatinib resistance in HCC.
Collapse
Affiliation(s)
- Nan Yi
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Lingyun Zhang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiangbo Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Jilei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China.
| | - Jian Gao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
6
|
Chen M, Liu X, Kong L, Yu Y, Zang J, Li X, Guo R, Zhang L, Liu Y. Efficacy assessment of glycyrrhetinic acid-modified liposomes loaded with doxorubicin hydrochloride and cucurbitine B for synergistic treatment of hepatocellular carcinoma. Int J Pharm 2025; 673:125360. [PMID: 39954971 DOI: 10.1016/j.ijpharm.2025.125360] [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: 11/26/2024] [Revised: 01/22/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is characterized by a high incidence rate, aggressive invasion and metastasis, and a significant postoperative recurrence rate. Targeted therapy plays a crucial role in the precise treatment of HCC. Studies have demonstrated that Glycyrrhetinic acid (GA) specific receptors are overexpressed on the surface of HCC cells. Doxorubicin hydrochloride (Dox), a widely used chemotherapy agent for anti-tumor treatment, but is associated with substantial toxic side effects. Cucurbitacin B (CuB) also demonstrates promising anti-tumor activity, but its poor water solubility and low bioavailability limit its clinical application. The combination of Dox and CuB can exert a synergistic effect, thereby enhancing the overall anti-tumor efficacy. Therefore, we have developed GA-modified liposomes loaded with Dox and CuB (GA-Dox/CuB-Lips) to achieve synergistic therapy for HCC. METHOD In this study, GA-Dox/CuB-Lips were prepared using the thin film dispersion method and ammonium sulfate gradient method. In vitro, we evaluated the cellular uptake and cytotoxicity of the liposomes, as well as their anti-tumor effects in inhibiting tumor proliferation, promoting tumor apoptosis, and suppressing invasion and metastasis. In vivo, the targeting properties of GA-Dox/CuB-Lips were assessed through in vivo imaging. A tumor growth curve was generated by establishing a heterotopic nude mouse model. Additionally, an in-situ HCC model was established and the anti-tumor effects of liposomes were evaluated using HE staining, histological analysis and immunofluorescence staining. RESULTS We successfully prepared GA-Dox/CuB-Lips with a smooth, spherical morphology and uniform distribution. Both drugs exhibited high encapsulation efficiency, significantly enhancing the solubility of CuB. In vitro, GA-Dox/CuB-Lips demonstrated excellent targeting properties and exerted cytotoxic effects on Hepa1-6 cells, effectively inhibiting tumor cell proliferation, invasion, and metastasis while promoting tumor cell apoptosis. In vivo, GA-Dox/CuB-Lips selectively targeted tumor sites, disrupted tumor structures, inhibited tumor growth and proliferation, and promoted apoptosis. CONCLUSION GA-Dox/CuB-Lips exhibited excellent anti-HCC activity and represent a promising therapeutic approach for the treatment of HCC.
Collapse
Affiliation(s)
- Muhan Chen
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Xinze Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China; Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Shenyang 110000, China
| | - Juan Zang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Xuetao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Ruibo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China.
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China.
| |
Collapse
|
7
|
Li W, Li Y, Wang M, Liu H, Hong G, Jiang L, Liu Z, Wu Y, Yuan L, Zhao X, He Z, Guo S, Xiao Y, Bi X, Xia M, Zou G, Zhang L, Gao J, Fu X. TNFAIP8L2 maintains hair cell function and regulates age-related hearing loss via mTORC1 signaling. Mol Ther 2025:S1525-0016(25)00218-7. [PMID: 40165373 DOI: 10.1016/j.ymthe.2025.03.046] [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: 10/29/2024] [Revised: 02/15/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
Age-related hearing loss (ARHL) is one of the most prevalent and complex disorders. Our previous study demonstrated that abnormal activation of mammalian target of rapamycin complex 1 (mTORC1) signaling in the cochlear neurosensory epithelium causes auditory hair cell (HC) damage and contributes to ARHL. However, the underlying mechanism of mTORC1 activation remains unclear. In this study, we identified tumor necrosis factor-alpha-induced protein 8-like 2 (TNFAIP8L2), an immune regulatory gene, as a potential candidate. To elucidate the effect of TNFAIP8L2 on mTORC1 signaling in the neurosensory epithelium and on hearing function, we generated a Tnfaip8l2-deficient (Tnfaip8l2-/-) mouse model. We discovered that Tnfaip8l2 deficiency led to features of oxidative stress in cochlear HCs and age-related hearing degeneration, exhibiting a similar phenotype to the mTORC1-over-activated Tsc1-cKO mice described previously. Furthermore, rapamycin, a well-known mTORC1 inhibitor, significantly mitigated the hearing dysfunction caused by Tnfaip8l2-deficiency. Mechanistically, we found that TNFAIP8L2 regulates mTORC1 signaling by simultaneously inhibiting the GTPase activity of Ras homolog enriched in brain (RHEB) and Ras-related C3 botulinum toxin substrate 1 (RAC1). Notably, both RHEB and RAC1 inhibitors alleviated the hearing phenotype observed in Tnfaip8l2-/- mice by inhibiting mTORC1 signaling. Collectively, our results provide insights into the activation of the mTORC1 pathway in aged mouse cochleae and positions TNFAIP8L2 as a valuable theoretical strategy.
Collapse
Affiliation(s)
- Wen Li
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yu Li
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Min Wang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Hao Liu
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Guodong Hong
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Luhan Jiang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Ziyi Liu
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yunhao Wu
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Liangjie Yuan
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xiaoxu Zhao
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zuhong He
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Siwei Guo
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yu Xiao
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xiuli Bi
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Ming Xia
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Guichang Zou
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Lining Zhang
- School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Jiangang Gao
- School of Life Science, Shandong University, Qingdao, Shandong 266237, China
| | - Xiaolong Fu
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
8
|
Shaikh S, Chary PS, Mehra NK. Tyrosine Kinase Inhibitor Lenvatinib Based Nano Formulations and Cutting-Edge Scale-Up Technologies in revolutionizing Cancer Therapy. ACS APPLIED BIO MATERIALS 2025; 8:1749-1784. [PMID: 40091597 DOI: 10.1021/acsabm.4c01527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Lenvatinib (LEN), a tyrosine kinase inhibitor, has emerged as a promising therapeutic agent for various solid tumors. Nevertheless, a number of constraints, including diminished bioavailability, incapacity to elicit localized inflammation, and inability to selectively accumulate at the tumor site, may impede the comprehensive exploitation of its versatile tyrosine kinase inhibitory capabilities. In order to achieve targeted delivery of LEN while also reducing its high dose used in conventional therapeutics, nanoformulation approaches can be adopted. The integration of LEN into various nanoformulations, such as nanoparticles, nanocrystals, high density lipoproteins (HDLs), liposomes, and micelles, is discussed, highlighting the advantages of these innovative approaches in a comparative manner; however, given that the current methods of nanoformulation synthesis employ toxic organic solvents and chemicals, there is an imperative need for exploring alternative, environmentally friendly approaches. The multifaceted effects of nanocarriers have rendered them profoundly applicable within the biomedical domain, serving as instrumental entities in various capacities such as vehicles for drug delivery and genetic material, diagnostic agents, facilitators of photothermal therapy, and radiotherapy. However, the scalability of these nanotechnological methodologies must be rigorously investigated and addressed to refine drug delivery mechanisms. This endeavor offers promising prospects for revolutionizing strategies in cancer therapeutics, thereby laying the foundation for future research in scale-up techniques in the pursuit of more effective and less toxic therapies for cancer.
Collapse
Affiliation(s)
- Samia Shaikh
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Padakanti Sandeep Chary
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| |
Collapse
|
9
|
Wu B, Cheng Y, Li L, Du Z, Liu Q, Tan X, Li X, Zhao G, Li E. Role of the sulfur-containing amino acid-ROS axis in cancer chemotherapeutic drug resistance. Drug Resist Updat 2025; 81:101238. [PMID: 40107045 DOI: 10.1016/j.drup.2025.101238] [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: 02/07/2025] [Revised: 03/10/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Chemotherapeutic drug resistance remains a major barrier to effective cancer treatment. Drug resistance could be driven in part by adaptive redox remodeling of cancer cells. Paradoxically, drug-resistant malignancies exhibit elevated reactive oxygen species (ROS), as well as amplified antioxidant defenses, which enable cancer cell survival under therapeutic stress. Central to this adaptation is glutathione (GSH), the predominant cellular antioxidant, whose synthesis relies on sulfur-containing amino acids (SAAs) - methionine and cysteine. This review delineates the metabolic interplay between methionine and cysteine in the transsulfuration pathway, highlighting their roles as precursors in GSH biosynthesis. We systematically summarize the key enzymes that drive GSH production and their contributions to resistance against platinum-based drugs and other chemotherapeutics. In addition to GSH synthesis, we summarize the roles of GSH antioxidant systems, including glutathione peroxidases (GPXs), peroxiredoxins (PRDXs), and thioredoxins (TRXs), which are critical in chemotherapeutic drug resistance through ROS scavenging. Recent advances reveal that targeting these enzymes, by pharmacologically inhibiting transsulfuration enzymes or disrupting GSH-dependent antioxidant cascades, can sensitize resistant cancer cells to ROS-mediated therapies. These findings not only clarify the mechanistic links between SAA metabolism and redox adaptation but also provide practical approaches to overcome chemotherapeutic drug resistance. By analyzing metabolic and redox vulnerabilities, this review highlights the therapeutic potential to restore chemosensitivity, offering new options in precision oncology medicine.
Collapse
Affiliation(s)
- Bingli Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong Province 515041, China; Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China.
| | - Yinwei Cheng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong Province 515041, China; Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Liyan Li
- Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province 518000, China
| | - Zepeng Du
- Department of Central Laboratory, Shantou Central Hospital, Shantou, Guangdong 515041, China
| | - Qianlou Liu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong Province 515041, China; Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xinyue Tan
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong Province 515041, China; Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong Province 515041, China; Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Guozhi Zhao
- Department of Urology Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong Province 515041, China; Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China.
| |
Collapse
|
10
|
Yang L, Tan W, Wang M, Wei Y, Xie Z, Wang Q, Zhang Z, Zhuang H, Ma X, Wang B, Jiang J, Chen Y, Shang C. circCCNY enhances lenvatinib sensitivity and suppresses immune evasion in hepatocellular carcinoma by serving as a scaffold for SMURF1 mediated HSP60 degradation. Cancer Lett 2025; 612:217470. [PMID: 39826668 DOI: 10.1016/j.canlet.2025.217470] [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: 07/11/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Lenvatinib is the standard first-line therapy for advanced hepatocellular carcinoma (HCC), but drug resistance significantly hampers its efficacy. Increasing evidence has shown that circular RNAs (circRNAs) play critical roles in HCC pathogenesis. However, the underlying mechanisms of lenvatinib sensitivity regulated by circRNAs remain largely unclear. The present study aims to identify circRNAs involved in lenvatinib resistance, as well as to elucidate the underlying mechanisms. High-throughput sequencing revealed that hsa_circ_0000235 (circCCNY) was downregulated in matched HCC tumor tissues and lenvatinib-resistant cells. Both in vitro and in vivo experiments revealed that downregulation of circCCNY could induce lenvatinib resistance in HCC cells. Subsequently, RNA pull-down, mass spectrometry, and RNA immunoprecipitation techniques were employed to investigate the interactions between circCCNY, HSP60, and the E3 ubiquitin ligase SMURF1. Briefly, circCCNY bounds to HSP60, subsequently leading to HSP60 ubiquitination and degradation through its interaction with the E3 ubiquitin ligase SMURF1. As a result, HSP60 degradation released Raf kinase inhibitor protein (RKIP), leading to the inactivation of the MAPK signaling pathway, and subsequently enhanced the anti-tumor effect of lenvatinib against HCC. Moreover, we also demonstrated that circCCNY could enhance CD8+ T-cell infiltration and suppress immune evasion through inhibiting the MAPK/c-Myc/PD-L1 signaling pathway. Our findings revealed that circCCNY enhances HCC sensitivity to lenvatinib and suppresses immune evasion by inhibiting the MAPK signaling pathway in HCC. This suggests that circCCNY could serve as a promising therapeutic target in HCC treatment and a potential biomarker for predicting HCC sensitivity to lenvatinib.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Phenylurea Compounds/pharmacology
- Phenylurea Compounds/therapeutic use
- Quinolines/pharmacology
- Quinolines/therapeutic use
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitin-Protein Ligases/genetics
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Animals
- Drug Resistance, Neoplasm/genetics
- Mice
- Cell Line, Tumor
- Tumor Escape/drug effects
- Gene Expression Regulation, Neoplastic
- Xenograft Model Antitumor Assays
- Male
- Mice, Nude
- Female
- Ubiquitination
Collapse
Affiliation(s)
- Lei Yang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui, China
| | - Wenliang Tan
- Center of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Min Wang
- Department of Pharmacy, Hainan General Hospital, Hainan Medical University, Haikou, 570311, Hai Nan, China
| | - Yingcheng Wei
- Department of Hepatopancreatobiliary Surgery, Shenshan Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Shanwei, 516621, Guangdong, China
| | - Zhiqin Xie
- Center of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Qingbin Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Ziyu Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Hongkai Zhuang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Xiaowu Ma
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Bingkun Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Jiahao Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Yajin Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Changzhen Shang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
11
|
Mu X, Pan L, Wang X, Liu C, Li Y, Cai Y, He Z. Development of a prognostic model for hepatocellular carcinoma based on microvascular invasion characteristic genes by spatial transcriptomics sequencing. Front Immunol 2025; 16:1529569. [PMID: 40051627 PMCID: PMC11882567 DOI: 10.3389/fimmu.2025.1529569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Microvascular invasion (MVI) is an independent risk factor for the recurrence and metastasis of hepatocellular carcinoma (HCC), associated with poor prognosis. Thus, MVI has significant clinical value for the treatment selection and prognosis assessment of patients with HCC. However, there is no reliable and precise method for assessing the postoperative prognosis of MVI patients. This study aimed to develop a new HCC prognosis prediction model based on MVI characteristic genes through spatial transcriptomics sequencing, distinguishing between high-risk and low-risk patients and evaluating patient prognosis. In this study, four MVI samples with different grades were selected for spatial transcriptomic sequencing to screen for MVI region-specific genes. On this basis, an HCC prognostic model was constructed using univariate Cox regression analysis, LASSO regression analysis, random survival forest, and stepwise multivariate Cox regression analysis methods. We constructed a 7-gene prognostic model based on MVI characteristic genes and demonstrated its applicability for predicting the prognosis of HCC patients in three external validation cohorts. Furthermore, our model showed superior predictive performance compared with three published HCC prediction prognostic models and could serve as an independent prognostic factor for HCC. Additionally, single nucleus RNA sequencing analysis and multiple immunofluorescence images revealed an increased proportion of macrophages in high-risk patient samples, suggesting that HCC tumor cells may promote HCC metastasis through MIF-CD74 cell interactions. To sum up, we have developed a 7-gene biomarker based on MVI that can predict the survival rate of HCC patients at different stages. This predictive model can be used to categorize into high- and low- risk groups, which is of great significance for the prognostic assessment and personalized treatment of HCC patients.
Collapse
Affiliation(s)
- Xiaolan Mu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lili Pan
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xicheng Wang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Changcheng Liu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Science and Technology Commission of Shanghai Municipality, Shanghai, China
| | - Yu Li
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yongchao Cai
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Science and Technology Commission of Shanghai Municipality, Shanghai, China
| | - Zhiying He
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Science and Technology Commission of Shanghai Municipality, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Municipal Education Commission, Shanghai, China
| |
Collapse
|
12
|
Chen R, Hu X, Huang Y, Jiang Y, Chen G, Shan Q, Xu X, Zheng S. Regulated Cell Death in Lenvatinib Resistance of Hepatocellular Carcinoma: from Molecular Mechanisms to Therapeutic Strategies. Int J Biol Sci 2025; 21:2012-2026. [PMID: 40083703 PMCID: PMC11900801 DOI: 10.7150/ijbs.107195] [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: 11/19/2024] [Accepted: 02/08/2025] [Indexed: 03/16/2025] Open
Abstract
Lenvatinib, a multi-target tyrosine kinase inhibitor (TKI), has been established as the first-line treatment for advanced hepatocellular carcinoma (HCC) because of its superior efficacy when in comparison with sorafenib. However, the inevitable development of drug resistance is a significant barrier to achieve a curative outcome and negatively impacts the prognosis. Therefore, it is imperative to delve into the mechanisms underlying lenvatinib resistance (LR) and to identify potential strategies for rational combination treatments. Regulated cell death (RCD) refers to the process by which cells undergo demise when the adaptive responses are insufficient to maintain homeostasis, and RCD takes a crucial part in the disease progression and response to therapeutic agents including TKI of cancer. Resisting cell death is one of the fundamental hallmarks and the major reasons contributing to drug resistance in cancer. Particularly, numerous studies have demonstrated that RCD (including apoptosis, autophagy, ferroptosis, cuproptosis and pyroptosis) plays a significant role in the emergence of LR in HCC. This article offers an in-depth review of recent discoveries concerning the mechanisms of LR in relation to RCD and proposes potential strategies to boost the effectiveness of lenvatinib by incorporating RCD modulators.
Collapse
Affiliation(s)
- Ronggao Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Xin Hu
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yingchen Huang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yao Jiang
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310059, China
| | - Guanrong Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiaonan Shan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Xiao Xu
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310059, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310000, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| |
Collapse
|
13
|
Wei Y, Yang L, Tang C, Zhuang H, Chen X, Ma X, Deng X, Chen Y, Tan W, Shang C. Lenvatinib inhibits cholangiocarcinoma progression by targeting the FGF19/PI3K/AKT signaling pathway. Apoptosis 2025; 30:185-196. [PMID: 39522105 DOI: 10.1007/s10495-024-02028-2] [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] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
Cholangiocarcinoma (CCA) is known for its high aggressiveness and dismal prognosis, whose effectiveness of systemic therapy remains limited. As a multi-target drug, lenvatinib has exhibited promising effects in many solid tumors. However, the therapeutic role of lenvatinib in CCA is rarely investigated. Here, the in vitro assays including EdU, colony formation, transwell, wound healing, and apoptosis analyses demonstrated that lenvatinib significantly inhibited the proliferation, migration, and invasion, while simultaneously inducing apoptosis of CCA cells. Mechanistically, lenvatinib downregulated the expression of FGF19 and inactivated the PI3K/AKT signaling pathway. Depletion of FGF19 enhanced the anti-tumor effects of lenvatinib, which was attributed to the inhibition of p-PI3K and p-AKT expression in CCA cells. In contrast, overexpression of FGF19 activated the PI3K/AKT signaling pathway, thereby impairing the inhibitory effects of lenvatinib against CCA. In addition, the AKT inhibitor, MK-2206, reinforced the lenvatinib-induced CCA inhibition. Notably, the in vivo experiment confirmed that the subcutaneous tumorigenicity of CCA cells in nude mice was weakened by lenvatinib. Lenvatinib markedly downregulated the expression of FGF19, p-AKT, Ki-67, vimentin, and VEGF in the xenograft tumor tissues. Collectively, these findings demonstrated that lenvatinib inhibits CCA progression by targeting the FGF19/PI3K/AKT signaling pathway. The present study provides novel experimental evidence for the potential clinical application of lenvatinib in CCA, which also highlights the promising role of targeting FGF19 in combined therapeutic approaches for CCA.
Collapse
Affiliation(s)
- Yingcheng Wei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
- Department of Hepatopancreatobiliary Surgery, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, 516621, Guangdong, China
| | - Lei Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Chenwei Tang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Hongkai Zhuang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Xinming Chen
- Department of Breast Surgery, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, 516621, Guangdong, China
| | - Xiaowu Ma
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Xuesong Deng
- Department of Hepatobiliary Surgery, Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Yajin Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Wenliang Tan
- Department of Hepatobiliary and Pancreatic Surgery, Medical Center of Digestive Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China.
| | - Changzhen Shang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
14
|
Wang D, Ma W, Zhang Y, Wang Y, Sun L, Jiang J, Jiao L, Li R, Zhang Y, Zhang M, Zhou Q. A versatile nanoplatform carrying cascade Pt nanozymes remodeling tumor microenvironment for amplified sonodynamic/chemo therapy of thyroid cancer. Biomaterials 2025; 313:122778. [PMID: 39213978 DOI: 10.1016/j.biomaterials.2024.122778] [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: 04/19/2024] [Revised: 08/06/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Thyroid cancer is increasing globally, with anaplastic thyroid carcinoma (ATC) being the most aggressive type and having a poor prognosis. Current clinical treatments for thyroid cancer present numerous challenges, including invasiveness and the necessity of lifelong medication. Furthermore, a significant portion of patients with ATC experience cancer recurrence and metastasis. To overcome this dilemma, we developed a pH-responsive biomimetic nanocarrier (CLP@HP-A) through the incorporation of Chlorin e6 (Ce6) and Lenvatinib (Len) within hollow polydopamine nanoparticles (HP) that were further modified with platinum nanoparticles (Pt), enabling synergistic chemotherapy and sonodynamic therapy. The CLP@HP-A nanocarriers exhibited specific binding with galectin-3 receptors, facilitating their internalization through receptor-mediated endocytosis for targeted drug delivery. Upon exposure to ultrasound (US) irradiation, Ce6 rapidly generated reactive oxygen species (ROS) to induce significant oxidative stress and trigger apoptosis in tumor cells. Additionally, Pt not only alleviated tumor hypoxia by catalyzing the conversion of H2O2 to oxygen (O2) but also augmented intracellular ROS levels through the production of hydroxyl radicals (•OH), thereby enhancing the efficacy of sonodynamic therapy. Moreover, Len demonstrated a potent cytotoxic effect on thyroid cancer cells through the induction of apoptosis. Transcriptomics analysis findings additionally corroborated that CLP@HP-A effectively triggered cancer cell apoptosis, thereby serving as a crucial mechanism for its cytotoxic effects. In conclusion, the integration of sonodynamic/chemo combination therapy with targeted drug delivery systems offers a novel approach to the management of malignant tumors.
Collapse
Affiliation(s)
- Dan Wang
- Department of Ultrasound, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Wenqi Ma
- Department of Ultrasound, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Yuanyuan Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yufeng Wang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Lei Sun
- Department of Ultrasound, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jue Jiang
- Department of Ultrasound, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Lianying Jiao
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Runqing Li
- Department of Radiology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China; Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Qi Zhou
- Department of Ultrasound, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
15
|
Ma X, Zhou K, Yan T, Hu L, Xie S, Zheng H, Tong Y, Zhang H, Wang Y, Gong Z, Chen C, Tian Y, Guo L, Lu R. Calpain 2 promotes Lenvatinib resistance and cancer stem cell traits via both proteolysis-dependent and independent approach in hepatocellular carcinoma. MOLECULAR BIOMEDICINE 2024; 5:74. [PMID: 39739077 PMCID: PMC11688263 DOI: 10.1186/s43556-024-00242-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
Lenvatinib, an approved first-line regimen, has been widely applied in hepatocellular carcinoma (HCC). However, clinical response towards Lenvatinib was limited, emphasizing the importance of understanding the underlying mechanism of its resistance. Herein, we employed integrated bioinformatic analysis to identify calpain-2 (CAPN2) as a novel key regulator for Lenvatinib resistance in HCC, and its expression greatly increased in both Lenvatinib-resistant HCC cell lines and clinical samples. Further in vitro and in vivo experiments indicated that knocking down CAPN2 greatly sensitized HCC cells to Lenvatinib treatment, while overexpression of CAPN2 achieved opposite effects in a Lenvatinib-sensitive HCC cell line. Interestingly, we observed a close relationship between CAPN2 expression and cancer stem cell (CSC) traits in HCC cells, evidenced by impaired sphere-forming and CSC-related marker expressions after CAPN2 knockdown, and verse vice. Mechanistically, we strikingly discovered that CAPN2 exerted its function by both enzyme-dependent and enzyme-independent manner simultaneously: activating β-Catenin signaling through its enzyme activity, and preventing GLI1/GLI2 degradation through direct binding to YWHAE in an enzyme-independent manner, which disrupting the association between YWHAE and GLI1/GLI2 to inhibit YWHAE-induced degradation of GLIs. Notably, further co-immunoprecipitation assays revealed that YWHAE could promote the protein stability of CAPN2 via recruiting a deubiquitinase COPS5 to prevent ubiquitination-induced degradation of CAPN2. In summary, our data demonstrated that CAPN2 promoted Lenvatinib resistance via both catalytic activity-dependent and -independent approaches. Reducing CAPN2 protein rather than inhibiting its activity might be a promising strategy to improve Lenvatinib treatment efficiency in HCC.
Collapse
Affiliation(s)
- Xiaolu Ma
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Kaixia Zhou
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Tianqing Yan
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Ling Hu
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Suhong Xie
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Hui Zheng
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Ying Tong
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Heng Zhang
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Yanchun Wang
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Zhiyun Gong
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Cuncun Chen
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Yanan Tian
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Lin Guo
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China.
| | - Renquan Lu
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| |
Collapse
|
16
|
Jia W, Xu B, Yu L, Feng Y, Wang J, Xu C, Liang L, Zhou Y, Ding W, Kong L. BAIAP2L2 promotes the malignancy of hepatocellular carcinoma via GABPB1-mediated reactive oxygen species imbalance. Cancer Gene Ther 2024; 31:1868-1883. [PMID: 39496939 PMCID: PMC11645275 DOI: 10.1038/s41417-024-00841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 11/06/2024]
Abstract
Hepatocellular carcinoma (HCC) is a common type of cancer worldwide and ranks as the fourth leading cause of cancer-related deaths. This research investigation identified an upregulation of BAI1-associated protein 2-like 2 (BAIAP2L2) in HCC tissues, which was found to be an independent prognostic factor for overall survival in HCC patients. BAIAP2L2 was observed to enhance cell proliferation, metastasis, stemness, cell cycle progression, and inhibit apoptosis in HCC. Mechanistically, NFκB1 was found to stimulate BAIAP2L2 transcription by directly binding to its promoter region. BAIAP2L2 interacts with GABPB1 to inhibit its ubiquitin-mediated degradation and promote its nuclear translocation. BAIAP2L2 inhibits the levels of reactive oxygen species (ROS) by regulating GABPB1, thereby promoting cancer properties in HCC and reducing the sensitivity of HCC to lenvatinib. In summary, this study elucidates the role and underlying mechanism of BAIAP2L2 in HCC, providing a potential biomarker and therapeutic target for this disease.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Apoptosis/genetics
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Proliferation/genetics
- Drug Resistance, Neoplasm/genetics
- GA-Binding Protein Transcription Factor/metabolism
- Gene Expression Regulation, Neoplastic
- Liver/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred BALB C
- Mice, Nude
- NF-kappa B p50 Subunit/metabolism
- Organoids
- Prognosis
- Promoter Regions, Genetic
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Proteolysis
- Reactive Oxygen Species/metabolism
- Spheroids, Cellular
- Up-Regulation
- Xenograft Model Antitumor Assays
- Female
- Middle Aged
- Kaplan-Meier Estimate
- Survival Rate
Collapse
Affiliation(s)
- Wenbo Jia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Bin Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Liang Yu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yanzhi Feng
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Jinyi Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Chao Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Litao Liang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Yongping Zhou
- Department of Hepatobiliary, Jiangnan University Medical Center, JUMC, Wuxi, Jiangsu Province, China.
| | - Wenzhou Ding
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China.
| | - Lianbao Kong
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China.
| |
Collapse
|
17
|
Kim S, Jeong N, Park J, Noh H, Lee JO, Yu SJ, Ku JL. Establishment and characterization of mouse metabolic dysfunction-associated steatohepatitis-related hepatocellular carcinoma organoids. Sci Rep 2024; 14:27460. [PMID: 39523389 PMCID: PMC11551198 DOI: 10.1038/s41598-024-78963-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a form of chronic liver inflammation associated with metabolic syndrome, such as obesity and a major cause of hepatocellular carcinoma (HCC). Multi-biotics, a soymilk fermented with lactic acid bacteria, are known to alleviate obesity by lowering lipid profile. This study aimed to establish and characterize mouse organoids derived from MASH-related HCC models to evaluate drug responses, particularly focusing on Lenvatinib resistance. Organoids were developed using mouse liver tissues subjected to a choline-deficient L-amino acid-defined high-fat diet (CDAHFD) to mimic MASH-related HCC. The study evaluated the effect of multi-biotics, a fermented product, on tumor regression and drug sensitivity. While multi-biotics did not reduce tumor burden, they enhanced the response to Lenvatinib. Additionally, repeated treatment with Lenvatinib led to the development of drug-resistant organoids. Transcriptomic analysis of these resistant organoids identified key pathways related to KRAS signaling, inflammation, and epithelial-mesenchymal transition (EMT), revealing potential targets for overcoming Lenvatinib resistance. This study provides valuable insights into MASH-related HCC progression and drug resistance, offering a model for further therapeutic research.
Collapse
Affiliation(s)
- Sumin Kim
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Korea
| | - Nahyun Jeong
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Korea
| | - Jeayeon Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hyojin Noh
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Ja Oh Lee
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Korea
| | - Su Jong Yu
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Ja-Lok Ku
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Cancer Research Institute, Seoul National University, Seoul, 03080, Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
18
|
Yang X, Zhang Q, Li D, Hu L, Wang Y, Yan X, Li Y, Wang Y, Zhang F, Shen J. A Multifunctional Nanodrug Increases the Therapeutic Sensitivity of Lenvatinib to Hepatocellular Carcinoma by Inhibiting the Stemness of Hepatic Cancer Stem Cells. Adv Healthc Mater 2024; 13:e2401398. [PMID: 39359011 DOI: 10.1002/adhm.202401398] [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: 04/18/2024] [Revised: 08/17/2024] [Indexed: 10/04/2024]
Abstract
Drug resistance resulting from diverse mechanisms including the presence of cancer stem cells (CSCs) is the main obstacle for improving therapeutic efficacy of lenvatinib in hepatocellular carcinoma (HCC). Herein, a nanomedicine (siCD24-Len-MnO@PLAP) is developed by incorporating manganese oxide (MnO), lenvatinib (Len), and siRNA against CD24 (siCD24) into micelles composed of methoxypolyethylene glycol (mPEG), poly-L-lysine (PLLys), and polyasparagyl(N-(2-Aminoethyl)piperidine) (PAsp(PIP)) triblock copolymer. The nanomedicine can respond to the tumor microenvironment (TME) to release lenvatinib, and produce Mn2+ and O2, accompanied by changes in nanoparticle charge, which facilitates cellular endocytosis of siCD24-loaded nanoparticles. The released siCD24 and lenvatinib synergistically reduces CD24 expression, resulting in a more pronounced inhibition of stemness of CSCs. In the mouse models of HCC using Huh7-derived CSCs and Hepa1-6-derived CSCs, the nanomedicine shows remarkable anti-cancer effect by enhancing the therapeutic effects of lenvatinib against HCC via reducing the expression level of CD24 and decreasing the expression of hypoxia inducible factor-1α (HIF-1α). Moreover, in situ production of paramagnetic Mn2+ from the nanomedicine serves as an excellent contrast agent for magnetic resonance imaging (MRI) to monitor the therapeutic process. This study demonstrates that this multifunctional MRI-visible siCD24- and lenvatinib-loaded nanodrug holds great potential in enhancing therapeutic sensitivity for HCC lenvatinib therapy.
Collapse
Affiliation(s)
- Xieqing Yang
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Qiaoyun Zhang
- College of Chemistry and Materials Science, Jinan University, No.855 Xingye Road East, Guangzhou, Guangdong, 510632, China
| | - Dongye Li
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Lanxin Hu
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu Wang
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xinyu Yan
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yunhua Li
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, No.855 Xingye Road East, Guangzhou, Guangdong, 510632, China
| | - Fang Zhang
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jun Shen
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| |
Collapse
|
19
|
Jin X, Lou X, Qi H, Zheng C, Li B, Siwu X, Liu R, Lv Q, Zhao A, Ruan J, Jiang M. NRF2 signaling plays an essential role in cancer progression through the NRF2-GPX2-NOTCH3 axis in head and neck squamous cell carcinoma. Oncogenesis 2024; 13:35. [PMID: 39333079 PMCID: PMC11437035 DOI: 10.1038/s41389-024-00536-z] [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: 04/05/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
The activation of nuclear factor erythroid 2-related factor 2 (NRF2) has been observed in various cancers. Yet its exact contribution to the development of head and neck squamous cell carcinoma (HNSCC) remains undetermined. We previously found that NRF2 signaling is critical for the differentiation of squamous basal progenitor cells, while disruption of NRF2 causes basal cell hyperplasia. In this study, we revealed a correlation between elevated NRF2 activity and poor outcomes in HNSCC patients. We demonstrated that NRF2 facilitates tumor proliferation, migration, and invasion, as evidenced by both in vitro and in vivo studies. Significantly, NRF2 augments the expression of the antioxidant enzyme GPX2, thereby enhancing the proliferative, migratory, and invasive properties of HNSCC cells. Activation of GPX2 is critical for sustaining cancer stem cells (CSCs) by up-regulating NOTCH3, a key driver of cancer progression. These results elucidate that NRF2 regulates HNSCC progression through the NRF2-GPX2-NOTCH3 axis. Our findings proposed that pharmacological targeting of the NRF2-GPX2-NOTCH3 axis could be a potential therapeutic approach against HNSCC.
Collapse
Affiliation(s)
- Xiaoye Jin
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Xiayuan Lou
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Haoxiang Qi
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| | - Chao Zheng
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Bo Li
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Xuerong Siwu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Ren Liu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Qiaoli Lv
- Institute of Cancer Research, Jiangxi Cancer Hospital, Nanchang, China
| | - An Zhao
- Institute of Cancer Research, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, China
| | - Ming Jiang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China.
| |
Collapse
|
20
|
Ye G, Ye M, Jin X. Roles of clinical application of lenvatinib and its resistance mechanism in advanced hepatocellular carcinoma (Review). Am J Cancer Res 2024; 14:4113-4171. [PMID: 39417171 PMCID: PMC11477829 DOI: 10.62347/ujvp4361] [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: 05/09/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Lenvatinib (LEN) is a multi-target TKI, which plays a pivotal role in the treatment of advanced hepatocellular carcinoma (HCC). The inevitable occurrence of drug resistance still prevents curative potential and is deleterious for the prognosis, and a growing body of studies is accumulating, which have devoted themselves to unveiling its underlying resistance mechanism and made some progress. The dysregulation of crucial signaling pathways, non-coding RNA and RNA modifications were proven to be associated with LEN resistance. A range of drugs were found to influence LEN therapeutic efficacy. In addition, the superiority of LEN combination therapy has been shown to potentially overcome the limitations of LEN monotherapy in a series of research, and a range of promising indicators for predicting treatment response and prognosis have been discovered in recent years. In this review, we summarize the latest developments in LEN resistance, the efficacy and safety of LEN combination therapy as well as associated indicators, which may provide new insight into its resistance as well as ideas in the treatment of advanced HCC.
Collapse
Affiliation(s)
- Ganghui Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
- Department of Radiation Oncology, Taizhou Central Hospital (Taizhou University Hospital)Taizhou 318000, Zhejiang, P. R. China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| |
Collapse
|
21
|
Qin Y, Han S, Yu Y, Qi D, Ran M, Yang M, Liu Y, Li Y, Lu L, Liu Y, Li Y. Lenvatinib in hepatocellular carcinoma: Resistance mechanisms and strategies for improved efficacy. Liver Int 2024; 44:1808-1831. [PMID: 38700443 DOI: 10.1111/liv.15953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024]
Abstract
Hepatocellular carcinoma (HCC), one of the most prevalent and destructive causes of cancer-related deaths worldwide, approximately 70% of patients with HCC exhibit advanced disease at diagnosis, limiting the potential for radical treatment. For such patients, lenvatinib, a long-awaited alternative to sorafenib for first-line targeted therapy, has become a key treatment. Unfortunately, despite some progress, the prognosis for advanced HCC remains poor because of drug resistance development. However, the molecular mechanisms underlying lenvatinib resistance and ways to relief drug resistance in HCC are largely unknown and lack of systematic summary; thus, this review not only aims to explore factors contributing to lenvatinib resistance in HCC, but more importantly, summary potential methods to conquer or mitigate the resistance. The results suggest that abnormal activation of pathways, drug transport, epigenetics, tumour microenvironment, cancer stem cells, regulated cell death, epithelial-mesenchymal transition, and other mechanisms are involved in the development of lenvatinib resistance in HCC and subsequent HCC progression. To improve the therapeutic outcomes of lenvatinib, inhibiting acquired resistance, combined therapies, and nano-delivery carriers may be possible approaches.
Collapse
Affiliation(s)
- Yongqing Qin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
| | - Shisong Han
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
| | - Yahan Yu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
| | - Ding Qi
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
| | - Mengnan Ran
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Mingqi Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
| | - Yanyan Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
| | - Yunyi Li
- Department of Nephrology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
| | - Yu Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
| | - Yong Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong, China
| |
Collapse
|
22
|
Glorieux C, Liu S, Trachootham D, Huang P. Targeting ROS in cancer: rationale and strategies. Nat Rev Drug Discov 2024; 23:583-606. [PMID: 38982305 DOI: 10.1038/s41573-024-00979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 07/11/2024]
Abstract
Reactive oxygen species (ROS) in biological systems are transient but essential molecules that are generated and eliminated by a complex set of delicately balanced molecular machineries. Disruption of redox homeostasis has been associated with various human diseases, especially cancer, in which increased ROS levels are thought to have a major role in tumour development and progression. As such, modulation of cellular redox status by targeting ROS and their regulatory machineries is considered a promising therapeutic strategy for cancer treatment. Recently, there has been major progress in this field, including the discovery of novel redox signalling pathways that affect the metabolism of tumour cells as well as immune cells in the tumour microenvironment, and the intriguing ROS regulation of biomolecular phase separation. Progress has also been made in exploring redox regulation in cancer stem cells, the role of ROS in determining cell fate and new anticancer agents that target ROS. This Review discusses these research developments and their implications for cancer therapy and drug discovery, as well as emerging concepts, paradoxes and future perspectives.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shihua Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Metabolic Innovation Center, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
23
|
Lin W, Wang X, Diao M, Wang Y, Zhao R, Chen J, Liao Y, Long Q, Meng Y. Promoting reactive oxygen species accumulation to overcome tyrosine kinase inhibitor resistance in cancer. Cancer Cell Int 2024; 24:239. [PMID: 38982494 PMCID: PMC11234736 DOI: 10.1186/s12935-024-03418-x] [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: 09/16/2023] [Accepted: 06/22/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND In tumor treatment, protein tyrosine kinase inhibitors (TKIs) have been extensively utilized. However, the efficacy of TKI is significantly compromised by drug resistance. Consequently, finding an effective solution to overcome TKI resistance becomes crucial. Reactive oxygen species (ROS) are a group of highly active molecules that play important roles in targeted cancer therapy including TKI targeted therapy. In this review, we concentrate on the ROS-associated mechanisms of TKI lethality in tumors and strategies for regulating ROS to reverse TKI resistance in cancer. MAIN BODY Elevated ROS levels often manifest during TKI therapy in cancers, potentially causing organelle damage and cell death, which are critical to the success of TKIs in eradicating cancer cells. However, it is noteworthy that cancer cells might initiate resistance pathways to shield themselves from ROS-induced damage, leading to TKI resistance. Addressing this challenge involves blocking these resistance pathways, for instance, the NRF2-KEAP1 axis and protective autophagy, to promote ROS accumulation in cells, thereby resensitizing drug-resistant cancer cells to TKIs. Additional effective approaches inducing ROS generation within drug-resistant cells and providing exogenous ROS stimulation. CONCLUSION ROS play pivotal roles in the eradication of tumor cells by TKI. Harnessing the accumulation of ROS to overcome TKI resistance is an effective and widely applicable approach.
Collapse
Affiliation(s)
- Wei Lin
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
| | - Mingxin Diao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
| | - Yangwei Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
| | - Rong Zhao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
| | - Jiaping Chen
- Department of Cardiothoracic Surgery, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, China
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China.
| | - Qinghong Long
- Department of Internal Medicine, Renmin Hospital, Wuhan University, Wuhan, 430022, China.
| | - Yunchong Meng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China.
| |
Collapse
|
24
|
Luo J, Yao Z, Liang W, Song D, Zeng H, Jiang Y, Bao Z, Zheng J, Ding Y. Mechanistic insights into 125I seed implantation therapy for Cholangiocarcinoma: focus on ROS-Mediated apoptosis and the role of GPX2. J Cancer Res Clin Oncol 2024; 150:324. [PMID: 38914724 PMCID: PMC11196350 DOI: 10.1007/s00432-024-05840-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/06/2024] [Indexed: 06/26/2024]
Abstract
OBJECTIVES Cholangiocarcinoma (CCA) is a rare tumor with a poor prognosis and poses significant therapeutic challenges. Herein, we investigated the mechanism of efficacy of 125I seed implantation therapy in CCA, focusing on the induction of reactive oxygen species (ROS)-mediated apoptosis and the involvement of glutathione peroxidase 2 (GPX2). MATERIALS AND METHODS Human cholangiocarcinoma cell lines QBC939 and RBE were purchased for in vitro studies. In vivo studies were performed using a rabbit VX2 CCA model. Apoptosis and proliferation were detected by TUNEL staining and clone formation, respectively. ROS generation was detected by dihydroethidium staining. Histological evaluation was performed by hematoxylin and eosin staining. Protein expression was determined by Western blotting and immunohistochemistry. RESULTS Our results demonstrate that 125I seeds effectively inhibited tumor growth in the rabbit VX2 tumor model and promoted the apoptosis of CCA cells in vitro in a dose-dependent manner. Molecular analyses indicate a marked increase in reactive oxygen species (ROS) levels following treatment with 125I seeds, suggesting the involvement of ROS-mediated apoptosis in the therapeutic mechanism. Furthermore, the downregulation of glutathione peroxidase 2 (GPX2) was observed, indicating its potential role in modulating ROS-mediated apoptosis in CCA. CONCLUSION 125I seed implantation therapy exerts therapeutic effects on CCA by inducing ROS-mediated apoptosis. The downregulation of GPX2 may contribute to enhanced ROS accumulation and apoptotic cell death. These findings provide mechanistic insights into the therapeutic potential of 125I seed implantation for CCA and highlight ROS-mediated apoptosis and GPX2 regulation as promising targets for further investigation and therapeutic intervention in this malignancy.
Collapse
Affiliation(s)
- Jun Luo
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang, 310022, China
| | - Zheng Yao
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang, 310022, China
| | - Weiren Liang
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang, 310022, China
| | - Danjun Song
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang, 310022, China
| | - Hui Zeng
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang, 310022, China
| | - Yi Jiang
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang, 310022, China
| | - Zhehan Bao
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang, 310022, China
| | - Jiaping Zheng
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang, 310022, China.
| | - Yinan Ding
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
25
|
Tao X, Cheng M, Huang X, Chen J, Zhou Y, Liu T, Zheng X, Shen N, Zhang Y, Luo P, He Q, Yan H, Huang P. Dabrafenib alleviates hepatotoxicity caused by lenvatinib via inhibiting the death receptor signaling pathway. Toxicol Lett 2024; 397:163-173. [PMID: 38754640 DOI: 10.1016/j.toxlet.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024]
Abstract
Lenvatinib is a multi-target inhibitor that exerts anti-tumor effects by inhibiting angiogenesis and is now commonly used as a first-line treatment for hepatocellular carcinoma. However, with the widespread use of lenvatinib, the problem of serious and fatal hepatotoxicity has become increasingly prominent. Currently, the mechanism behind this toxicity is not yet understood, and as a result, there is a lack of safe and effective intervention strategies with minimal side effects. Here, we established the model of lenvatinib-induced liver injury in vivo and in vitro and found that lenvatinib caused hepatotoxicity by inducing apoptosis. Further mechanistic studies in cellular models revealed that lenvatinib upregulated death receptor signaling pathway, which activated the downstream effector Caspase-8, and ultimately led to apoptosis. Meanwhile, lenvatinib-induced apoptosis was associated with ROS generation and DNA damage. In addition, after screening marketed drugs and natural products in combination with cellular modeling, we identified a potential co-administered drug, dabrafenib, which could alleviate lenvatinib-induced hepatotoxicity. Further mechanistic studies revealed that dabrafenib attenuated lenvatinib-induced hepatotoxicity by inhibiting the activation of the death receptor signaling pathway. Subsequently, cancer cell proliferation assays confirmed that dabrafenib did not antagonize the antitumor effects of lenvatinib. In conclusion, our results validate that apoptosis caused by the death receptor signaling pathway is the key cause of lenvatinib-induced hepatotoxicity, and dabrafenib alleviates lenvatinib-induced hepatotoxicity by inhibiting this pathway.
Collapse
Affiliation(s)
- Xinyu Tao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Mengting Cheng
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiangliang Huang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiajia Chen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yunfang Zhou
- The Laboratory of Clinical Pharmacy, the Sixth Affiliated Hospital of Wenzhou Medical University, the People's Hospital of Lishui, Lishui, Zhejiang 323020, China
| | - Ting Liu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; International Center for Medical Translation and Innovation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaochun Zheng
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Nonger Shen
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang 310014, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Pharmacology and Toxicology, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang 310018, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Ping Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
26
|
Chen X, Zhu N, Wu Y, Zhang Y, Zhang Y, Jin K, Zhou Z, Chen G, Wang J. Withaferin A, a natural thioredoxin reductase 1 (TrxR1) inhibitor, synergistically enhances the antitumor efficacy of sorafenib through ROS-mediated ER stress and DNA damage in hepatocellular carcinoma cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155317. [PMID: 38537439 DOI: 10.1016/j.phymed.2023.155317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/22/2023] [Accepted: 12/25/2023] [Indexed: 05/01/2024]
Abstract
BACKGROUND Sorafenib (Sora), a multi-target tyrosine kinase inhibitor, is widely recognized as a standard chemotherapy treatment for advanced hepatocellular carcinoma (HCC). However, drug resistance mechanisms hinder its anticancer efficacy. Derived from Withania somnifera, Withaferin A (WA) exhibits remarkable anti-tumor properties as a natural bioactive compound. This study aimed to examine the mechanisms that underlie the impacts of Sora and WA co-treatment on HCC. METHODS Cell proliferation was evaluated through colony formation and MTT assays. Flow cytometry was employed to determine cellular apoptosis and reactive oxygen species (ROS) levels. The evaluation of apoptosis-related protein levels, DNA damage, and endoplasmic reticulum stress was conducte utilizing IHC staining and western blotting. Moreover, the caspase inhibitor Z-VAD-FMK, ATF4 siRNA, ROS scavenger N-acetyl cysteine (NAC), and TrxR1 shRNA were used to elucidate the underlying signaling pathways. To validate the antitumor effects of Sora/WA co-treatment, in vivo experiments were ultimately executed using Huh7 xenografts. RESULTS Sora/WA co-treatment demonstrated significant synergistic antitumor impacts both in vivo and in vitro. Mechanistically, the enhanced antitumor impact of Sora by WA was achieved through the inhibition of TrxR1 activity, resulting in ROS accumulation. Moreover, ROS generation induced the activation of DNA damage and endoplasmic reticulum (ER) stress pathways, eventually triggering cellular apoptosis. Pre-treatment with the antioxidant NAC significantly inhibited ROS generation, ER stress, DNA damage, and apoptosis induced by Sora/WA co-treatment. Additionally, the inhibition of ATF4 by small interfering RNA (siRNA) attenuated Sora/WA co-treatment-induced apoptosis. In vivo, Sora/WA co-treatment significantly suppressed tumor growth in HCC xenograft models and decreased TrxR1 activity in tumor tissues. CONCLUSION Our study suggests that WA synergistically enhances the antitumor effect of Sora, offering promising implications for evolving treatment approaches for HCC.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacology, School of Medicine, Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China
| | - Ning Zhu
- Municipal Hospital Affiliated to Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China
| | - Yajie Wu
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China
| | - Ye Zhang
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China
| | - Yuxuan Zhang
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China
| | - Kaiwen Jin
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China
| | - Zhiyi Zhou
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China
| | - Guang Chen
- Department of Pharmacology, School of Medicine, Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China
| | - Jiabing Wang
- Municipal Hospital Affiliated to Taizhou University, Taizhou, Jiaojiang 318000, Zhejiang, China.
| |
Collapse
|
27
|
Zhang C, Wang L, Qin L, Luo Y, Wen Z, Vignon AS, Zheng C, Zhu X, Chu H, Deng S, Hong L, Zhang J, Yang H, Zhang J, Ma Y, Wu G, Sun C, Liu X, Pu L. Overexpression of GPX2 gene regulates the development of porcine preadipocytes and skeletal muscle cells through MAPK signaling pathway. PLoS One 2024; 19:e0298827. [PMID: 38722949 PMCID: PMC11081289 DOI: 10.1371/journal.pone.0298827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 01/30/2024] [Indexed: 05/13/2024] Open
Abstract
Glutathione peroxidase 2 (GPX2) is a selenium-dependent enzyme and protects cells against oxidative damage. Recently, GPX2 has been identified as a candidate gene for backfat and feed efficiency in pigs. However, it is unclear whether GPX2 regulates the development of porcine preadipocytes and skeletal muscle cells. In this study, adenoviral gene transfer was used to overexpress GPX2. Our findings suggest that overexpression of GPX2 gene inhibited proliferation of porcine preadipocytes. And the process is accompanied by the reduction of the p-p38. GPX2 inhibited adipogenic differentiation and promoted lipid degradation, while ERK1/2 was reduced and p-p38 was increased. Proliferation of porcine skeletal muscle cells was induced after GPX2 overexpression, was accompanied by activation in JNK, ERK1/2, and p-p38. Overexpression methods confirmed that GPX2 has a promoting function in myoblastic differentiation. ERK1/2 pathway was activated and p38 was suppressed during the process. This study lays a foundation for the functional study of GPX2 and provides theoretical support for promoting subcutaneous fat reduction and muscle growth.
Collapse
Affiliation(s)
- Chunguang Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Lei Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Department of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Lei Qin
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Yunyan Luo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Zuochen Wen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Akpaca Samson Vignon
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Chunting Zheng
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Xueli Zhu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Han Chu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Shifan Deng
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Liang Hong
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin modern Tianjiao Agricultural Technology Co, LTD, Tianjin Key Laboratory of Green Ecological Feed, Tianjin, China
| | - Jianbin Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin modern Tianjiao Agricultural Technology Co, LTD, Tianjin Key Laboratory of Green Ecological Feed, Tianjin, China
| | - Hua Yang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Jianbo Zhang
- State Key Laboratory of Plateau Ecology and Agriculture, Department of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Yuhong Ma
- State Key Laboratory of Plateau Ecology and Agriculture, Department of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Guofang Wu
- State Key Laboratory of Plateau Ecology and Agriculture, Department of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Chao Sun
- Tianjin modern Tianjiao Agricultural Technology Co, LTD, Tianjin Key Laboratory of Green Ecological Feed, Tianjin, China
| | - Xin Liu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lei Pu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin modern Tianjiao Agricultural Technology Co, LTD, Tianjin Key Laboratory of Green Ecological Feed, Tianjin, China
| |
Collapse
|
28
|
Xue W, Wang T, Tian WJ, Pang SQ, Zhang HF, Jia WD. NQO1 Mediates Lenvatinib Resistance by Regulating ROS-induced Apoptosis in Hepatocellular Carcinoma. Curr Med Sci 2024; 44:168-179. [PMID: 38217831 DOI: 10.1007/s11596-023-2804-8] [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: 05/30/2023] [Accepted: 09/19/2023] [Indexed: 01/15/2024]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is the third leading cause of cancer-associated death worldwide. As a first-line drug for advanced HCC treatment, lenvatinib faces a significant hurdle due to the development of both intrinsic and acquired resistance among patients, and the underlying mechanism remains largely unknown. The present study aims to identify the pivotal gene responsible for lenvatinib resistance in HCC, explore the potential molecular mechanism, and propose combinatorial therapeutic targets for HCC management. METHODS Cell viability and colony formation assays were conducted to evaluate the sensitivity of cells to lenvatinib and dicoumarol. RNA-Seq was used to determine the differences in transcriptome between parental cells and lenvatinib-resistant (LR) cells. The upregulated genes were analyzed by GO and KEGG analyses. Then, qPCR and Western blotting were employed to determine the relative gene expression levels. Afterwards, the intracellular reactive oxygen species (ROS) and apoptosis were detected by flow cytometry. RESULTS PLC-LR and Hep3B-LR were established. There was a total of 116 significantly upregulated genes common to both LR cell lines. The GO and KEGG analyses indicated that these genes were involved in oxidoreductase and dehydrogenase activities, and reactive oxygen species pathways. Notably, NAD(P)H:quinone oxidoreductase 1 (NQO1) was highly expressed in LR cells, and was involved in the lenvatinib resistance. The high expression of NQO1 decreased the production of ROS induced by lenvatinib, and subsequently suppressed the apoptosis. The combination of lenvatinib and NQO1 inhibitor, dicoumarol, reversed the resistance of LR cells. CONCLUSION The high NQO1 expression in HCC cells impedes the lenvatinib-induced apoptosis by regulating the ROS levels, thereby promoting lenvatinib resistance in HCC cells.
Collapse
Affiliation(s)
- Wei Xue
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ting Wang
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Wen-Jing Tian
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Si-Qi Pang
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hua-Feng Zhang
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China.
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Wei-Dong Jia
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
29
|
Brzozowa-Zasada M, Ianaro A, Piecuch A, Michalski M, Matysiak N, Stęplewska K. Immunohistochemical Expression of Glutathione Peroxidase-2 (Gpx-2) and Its Clinical Relevance in Colon Adenocarcinoma Patients. Int J Mol Sci 2023; 24:14650. [PMID: 37834097 PMCID: PMC10572251 DOI: 10.3390/ijms241914650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
Glutathione peroxidase 2 (Gpx-2) is a selenoenzyme with antioxidant capabilities that may play a role in cancer development. Hence, we investigated the immunohistochemical expression of Gpx-2 protein in colon adenocarcinoma samples derived from patients with colon adenocarcinoma who did not receive any form of treatment prior to the surgical procedure. The associations between the immunohistochemical expression of Gpx-2 and clinical parameters were analysed using the Chi2 test and Fisher's exact test. A Kaplan-Meier analysis and the log-rank test were used to verify the relationship between the intensity of Gpx-2 expression and the 5-year survival rate of patients. In total, 101 (80.80%) samples had strong Gpx-2 protein expression and 24 (19.20%) samples were characterized with low expression. The high expression of Gpx-2 was correlated with the histological grade of the tumour (p < 0.001), PCNA immunohistochemical expression (p < 0.001), depth of invasion (p = 0.001) and angioinvasion (p < 0.001). We can conclude that high expression of Gpx-2 is correlated with reduced survival of colon adenocarcinoma patients (log-rank, p < 0.001).
Collapse
Affiliation(s)
- Marlena Brzozowa-Zasada
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Angela Ianaro
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
| | - Adam Piecuch
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Marek Michalski
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Natalia Matysiak
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Katarzyna Stęplewska
- Department of Pathology, Institute of Medical Sciences, University of Opole, 45-052 Opole, Poland
| |
Collapse
|
30
|
Chen W, Ruan M, Zou M, Liu F, Liu H. Clinical Significance of Non-Coding RNA Regulation of Programmed Cell Death in Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:4187. [PMID: 37627215 PMCID: PMC10452865 DOI: 10.3390/cancers15164187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a widely prevalent and malignantly progressive tumor. Most patients are typically diagnosed with HCC at an advanced stage, posing significant challenges in the execution of curative surgical interventions. Non-coding RNAs (ncRNAs) represent a distinct category of RNA molecules not directly involved in protein synthesis. However, they possess the remarkable ability to regulate gene expression, thereby exerting significant regulatory control over cellular processes. Notably, ncRNAs have been implicated in the modulation of programmed cell death (PCD), a crucial mechanism that various therapeutic agents target in the fight against HCC. This review summarizes the clinical significance of ncRNA regulation of PCD in HCC, including patient diagnosis, prognosis, drug resistance, and side effects. The aim of this study is to provide new insights and directions for the diagnosis and drug treatment strategies of HCC.
Collapse
Affiliation(s)
| | | | | | - Fuchen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai 200438, China; (W.C.); (M.R.)
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai 200438, China; (W.C.); (M.R.)
| |
Collapse
|
31
|
Suzuki S, Gi M, Fujioka M, Kakehashi A, Wanibuchi H. Dimethylarsinic acid induces bladder carcinogenesis via the amphiregulin pathway. Toxicol Lett 2023; 384:128-135. [PMID: 37567419 DOI: 10.1016/j.toxlet.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/27/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Dimethylarsinic acid (DMA) is a major metabolite in the urine of humans and rats exposed to inorganic arsenicals, and is reported to induce rat bladder carcinogenesis. In the present study, we focused on early pathways of carcinogenesis triggered by DMA that were also active in tumors. RNA expression in the bladder urothelium of rats treated with 0 and 200 ppm DMA in the drinking water for 4 weeks and in bladder tumors of rats treated with 200 ppm DMA for 2 years was initially examined using microarray analysis and Ingenuity Pathway Analysis (IPA). Expression of 160 genes was altered in both the urothelium of rats treated for 4 weeks with DMA and in DMA-induced tumors. IPA associated 36 of these genes with liver tumor diseases. IPA identified the amphiregulin (Areg)-regulated pathway as a Top Regulator Effects Network. Therefore, we focused on Areg and 6 of its target genes: cyclin A2, centromere protein F, marker of proliferation Ki-67, protein regulator of cytokinesis 1, ribonucleotide reductase M2, and topoisomerase II alpha. We confirmed high mRNA expression of Areg and its 6 target genes in both the urothelium of rats treated for 4 weeks with DMA and in DMA-induced tumors. RNA interference of human amphiregulin (AREG) expression in human urinary bladder cell lines T24 and UMUC3 decreased expression of AREG and its 6 target genes and decreased cell proliferation. These data suggest that Areg has an important role in DMA-induced rat bladder carcinogenesis.
Collapse
Affiliation(s)
- Shugo Suzuki
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Min Gi
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan; Department of Environmental Risk Assessment, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Masaki Fujioka
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Anna Kakehashi
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan.
| |
Collapse
|