1
|
Li Y, Chen Y, Zhang Y, Fang Y, Wu L, Zhao Y, Wang D, Qiao X. Integrating multi-omics techniques and in vitro experiments reveals that GLRX3 regulates the immune microenvironment and promotes hepatocellular carcinoma cell proliferation and invasion through iron metabolism pathways. Front Immunol 2024; 15:1496886. [PMID: 39654899 PMCID: PMC11625766 DOI: 10.3389/fimmu.2024.1496886] [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: 09/15/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a common malignancy worldwide, and its development is closely related to abnormalities in iron metabolism. This study aims to systematically analyze changes in iron metabolism in the tumor microenvironment of HCC using single-cell sequencing technology, and investigate the potential mechanisms by which iron metabolism regulation affects the survival of liver cancer patients. Materials and methods Single-cell sequencing data from hepatocellular carcinoma patients were obtained from the GEO database. By iron metabolism genomic scoring, we assessed differences in iron metabolism levels in hepatocellular carcinoma samples. By cell communication analysis as well as GO and KEGG enrichment analysis, we determined the functional role of iron metabolism in different cell types. We used survival analysis and Kaplan-Meier curves to assess the impact of iron metabolism levels on patient prognosis. In addition, we identified and analyzed the expression profile of the GLRX3 gene, investigated its key regulatory role in iron metabolism, and validated its clinical value as a prognostic marker. Finally, we explored the effect of GLRX3 on hepatocellular carcinoma phenotype by in vitro experiments such as PCR, transwell, CCK8, and wound healing assay. Results Bioinformatics results and experimental validation confirmed the dysregulation of iron metabolism in the development of hepatocellular carcinoma, revealing iron's regulatory influence across various cell types. Additionally, GLRX3 was identified as a key regulatory factor in iron metabolism, and the mechanism by which GLRX3 regulates tumor cell proliferation and immune evasion was determined. Furthermore, experiments verified GLRX3's role in facilitating tumor cell proliferation and invasion. Conclusion This study highlights the critical role of iron metabolism in the progression of hepatocellular carcinoma, particularly the regulatory mechanism of the GLRX3 gene in tumor cell proliferation and immune evasion. Iron metabolism abnormalities are not only drivers of liver cancer development but also key indicators of patient prognosis.
Collapse
Affiliation(s)
- Yang Li
- Department of General Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Yuan Chen
- Department of General Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Zhang
- School of Mechanical Engineering, Taiyuan University of Science and Technology, Taiyuan, China
| | - Yunsheng Fang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an, China
| | - Ling Wu
- Tumor Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Ying Zhao
- Department of General Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Danqiong Wang
- Department of General Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Xiaoyuan Qiao
- Department of Comprehensive Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Du H, Hou L, Yu H, Zhang F, Tong K, Wu X, Zhang Z, Liu K, Miao X, Guo W, Guo J, Kong Y. Enhancer of Zeste Homolog 2 Protects Mucosal Melanoma from Ferroptosis via the KLF14-SLC7A11 Signaling Pathway. Cancers (Basel) 2024; 16:3660. [PMID: 39518098 PMCID: PMC11545276 DOI: 10.3390/cancers16213660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Mucosal melanoma (MM) is epidemiologically, biologically, and molecularly distinct from cutaneous melanoma. Current treatment strategies have failed to significantly improve the prognosis for MM patients. This study aims to identify therapeutic targets and develop combination strategies by investigating the mechanisms underlying the tumorigenesis and progression of MM. METHODS We analyzed the copy number amplification of enhancer of zeste homolog 2 (EZH2) in 547 melanoma patients and investigated its correlation with clinical prognosis. Utilizing cell lines, organoids, and patient-derived xenograft models, we assessed the impact of EZH2 on cell proliferation and sensitivity to ferroptosis. Further, we explored the mechanisms of ferroptosis resistance associated with EZH2 by conducting RNA sequencing and chromatin immunoprecipitation sequencing. RESULTS EZH2 copy number amplification was closely associated with malignant phenotype and poor prognosis in MM patients. EZH2 was essential for MM cell proliferation in vitro and in vivo. Moreover, genetic perturbation of EZH2 rendered MM cells sensitized to ferroptosis. Combination treatment of EZH2 inhibitor with ferroptosis inducer significantly inhibited the growth of MM. Mechanistically, EZH2 inhibited the expression of Krüpple-Like factor 14 (KLF14), which binds to the promoter of solute carrier family 7 member 11 (SLC7A11) to repress its transcription. Loss of EZH2 therefore reduced the expression of SLC7A11, leading to reduced intracellular SLC7A11-dependent glutathione synthesis to promote ferroptosis. CONCLUSION Our findings not only establish EZH2 as a biomarker for MM prognosis but also highlight the EZH2-KLF14-SLC7A11 axis as a potential target for MM treatment.
Collapse
Affiliation(s)
- Haizhen Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| | - Lijie Hou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| | - Huan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Fenghao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| | - Ke Tong
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | - Xiaowen Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| | - Ziyi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| | - Kaiping Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| | - Xiangguang Miao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| | - Wenhui Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; (H.D.); (L.H.)
| |
Collapse
|
3
|
Ye H, Ding X, Lv X, Du Y, Guo R, Qiu J, Li R, Cao L. KLF14 directly downregulates the expression of GPX4 to exert antitumor effects by promoting ferroptosis in cervical cancer. J Transl Med 2024; 22:923. [PMID: 39390559 PMCID: PMC11465826 DOI: 10.1186/s12967-024-05714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Cervical cancer is the fourth leading cause of cancer-related death among women worldwide, and effective therapeutic strategies for its treatment are limited. Recent studies have indicated that ferroptosis, a form of regulated cell death, is a promising therapeutic strategy. KLF14 has been shown to regulate both cell proliferation and apoptosis in cervical cancer. However, its role in modulating lipid peroxidation and ferroptosis remains largely unexplored and enigmatic. METHODS SiHa and HeLa cells were transduced with lentiviral vectors to overexpress KLF14. Protein levels were analyzed via western blotting and immunohistochemistry (IHC). LDH assays, calcein-AM/propidium iodide (PI) staining, and generation of cell growth curves using a real-time cell analysis (RTCA) system were used to detect cell damage and proliferation. Cellular ROS, lipid ROS, transmission electron microscopy (TEM), and Fe2+ assays and a xenograft mouse model were used to measure the level of ferroptosis. Proteomics combined with bioinformatics methods was used to screen target genes regulated by KLF14, and CUT&Tag and dual-luciferase assays confirmed the repression of GPX4 by KLF14 via direct binding to its promoter. RESULTS KLF14 is abnormally expressed in various tumors and downregulated in cervical cancer. Overexpression of KLF14 induced ferroptosis and inhibited cell proliferation in vitro as well as xenograft tumorigenicity in vivo. Mechanistic studies revealed that KLF14 binds to the promoter of GPX4, suppressing its transcriptional activity and thereby decreasing its expression, which contributes to the induction of ferroptosis. Truncation and point mutation analyses of the GPX4 promoter revealed multiple binding sites for KLF14 within the - 1000 bp to + 35 bp region, which are responsible for its inhibitory effect on GPX4 transcription. Additionally, deletion of the zinc finger motif in KLF14 abolished its inhibitory effect on GPX4 promoter activity and cell proliferation. CONCLUSION Our data revealed a previously unidentified function of KLF14 in promoting ferroptosis, which results in the suppression of cell proliferation. Mechanistically, we revealed a novel regulatory mechanism by which KLF14 targets GPX4. These findings suggest a novel strategy to induce ferroptosis through the targeting of KLF14 in human cervical cancer cells.
Collapse
Affiliation(s)
- Hui Ye
- Oncology Department, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China
- Blood Center of Shandong Province, Jinan, 250014, China
| | - XuChao Ding
- Department of Blood Transfusion, Binzhou People's Hospital, Binzhou, 256601, China
| | - XinRan Lv
- Shandong Public Health Clinical Center, Shandong University, Shandong, 250013, China
| | - Ying Du
- Department of Laboratory Medicine, Linyi Peoples' Hospital, Linyi, 276003, China
| | - Rui Guo
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Jin Qiu
- Oncology Department, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - RuoNan Li
- Oncology Department, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - LiLi Cao
- Oncology Department, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China.
- Oncology Department, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China.
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Laboratory of Clinical Immunology and Translational Medicine, Shandong Provincial University, Jinan, 250014, China.
- Shandong Lung Cancer Institute, Jinan, 250014, China.
| |
Collapse
|
4
|
Xie Z, Che Y, Huang G, Su Z, Lin J, Zheng G, Ye G, Yu W, Li J, Wu Y, Shen H. Iron-dependent KDM4D activity controls the quiescence-activity balance of MSCs via the PI3K-Akt-Foxo1 pathway. Cell Mol Life Sci 2024; 81:360. [PMID: 39158700 PMCID: PMC11335281 DOI: 10.1007/s00018-024-05376-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/01/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024]
Abstract
Iron deficiency is a prevalent nutritional deficit associated with organ damage and dysfunction. Recent research increasingly associates iron deficiency with bone metabolism dysfunction, although the precise underlying mechanisms remain unclear. Some studies have proposed that iron-dependent methylation-erasing enzyme activity regulates cell proliferation and differentiation under physiological or pathological conditions. However, it remains uncertain whether iron deficiency inhibits the activation of quiescent mesenchymal stem cells (MSCs) by affecting histone demethylase activity. In our study, we identified KDM4D as a key player in the activation of quiescent MSCs. Under conditions of iron deficiency, the H3K9me3 demethylase activity of KDM4D significantly decreased. This alteration resulted in increased heterochromatin with H3K9me3 near the PIK3R3 promoter, suppressing PIK3R3 expression and subsequently inhibiting the activation of quiescent MSCs via the PI3K-Akt-Foxo1 pathway. Iron-deficient mice displayed significantly impaired bone marrow MSCs activation and decreased bone mass compared to normal mice. Modulating the PI3K-Akt-Foxo1 pathway could reverse iron deficiency-induced bone loss.
Collapse
Affiliation(s)
- Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Yunshu Che
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Department of Orthopedics Surgery, Suzhou Municipal Hospital/The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China
| | - Guo Huang
- Department of Rheumatology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Zepeng Su
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Jiajie Lin
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Guiwen Ye
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Wenhui Yu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Jinteng Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| |
Collapse
|
5
|
Yu X, Guo Q, Zhang H, Wang X, Han Y, Yang Z. Hypoxia-inducible factor-1α can reverse the Adriamycin resistance of breast cancer adjuvant chemotherapy by upregulating transferrin receptor and activating ferroptosis. FASEB J 2024; 38:e23876. [PMID: 39120539 DOI: 10.1096/fj.202401119r] [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: 05/18/2024] [Revised: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Breast cancer is a common malignant tumor in women. Ferroptosis, a programmed cell death pathway, is closely associated with breast cancer and its resistance. The transferrin receptor (TFRC) is a key factor in ferroptosis, playing a crucial role in intracellular iron accumulation and the occurrence of ferroptosis. This study investigates the influence and significance of TFRC and its upstream transcription factor hypoxia-inducible factor-1α (HIF1α) on the efficacy of neoadjuvant therapy in breast cancer. The differential gene obtained from clinical samples through genetic sequencing is TFRC. Bioinformatics analysis revealed that TFRC expression in breast cancer was significantly greater in breast cancer tissues than in normal tissues, but significantly downregulated in Adriamycin (ADR)-resistant tissues. Iron-responsive element-binding protein 2 (IREB2) interacts with TFRC and participates in ferroptosis. HIF1α, an upstream transcription factor, positively regulates TFRC. Experimental results indicated higher levels of ferroptosis markers in breast cancer tissue than in normal tissue. In the TAC neoadjuvant regimen-sensitive group, iron ion (Fe2+) and malondialdehyde (MDA) levels were greater than those in the resistant group (all p < .05). Expression levels of TFRC, IREB2, FTH1, and HIF1α were higher in breast cancer tissue compared to normal tissue. Additionally, the expression of the TFRC protein in the TAC neoadjuvant regimen-sensitive group was significantly higher than that in the resistant group (all p < .05), while the difference in the level of expression of IREB2 and FTH1 between the sensitive and resistant groups was not significant (p > .05). The dual-luciferase assay revealed that HIF1α acts as an upstream transcription factor of TFRC (p < .05). Overexpression of HIF1α in ADR-resistant breast cancer cells increased TFRC, Fe2+, and MDA content. After ADR treatment, the cell survival rate decreased significantly, and ferroptosis could be reversed by the combined application of Fer-1 (all p < .05). In conclusion, ferroptosis and chemotherapy resistance are correlated in breast cancer. TFRC is a key regulatory factor influenced by HIF1α and is associated with chemotherapy resistance. Upregulating HIF1α in resistant cells may reverse resistance by activating ferroptosis through TFRC overexpression.
Collapse
MESH Headings
- Female
- Humans
- Middle Aged
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Chemotherapy, Adjuvant/methods
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Drug Resistance, Neoplasm
- Ferroptosis/drug effects
- Gene Expression Regulation, Neoplastic
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- MCF-7 Cells
- Receptors, Transferrin/metabolism
- Receptors, Transferrin/genetics
- Up-Regulation
Collapse
Affiliation(s)
- Xiaojie Yu
- Department of Thyroid Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Qingqun Guo
- Department of Thyroid Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Haojie Zhang
- Department of Thyroid Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Xiaohong Wang
- Department of Breast Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Yong Han
- Department of Thyroid Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Zhenlin Yang
- Department of Thyroid Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| |
Collapse
|
6
|
Wu J, Shi Y, Zhou M, Chen M, Ji S, Liu X, Zhou M, Xia R, Zheng X, Wang W. Nutrient vitamins enabled metabolic regulation of ferroptosis via reactive oxygen species biology. Front Pharmacol 2024; 15:1434088. [PMID: 39092216 PMCID: PMC11291210 DOI: 10.3389/fphar.2024.1434088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Vitamins are dietary components necessary for cellular metabolic balance, especially redox homeostasis; deficient or excessive supply may give rise to symptoms of psychiatric disorders. Exploring the nutritional and metabolic pathways of vitamins could contribute to uncovering the underlying pathogenesis of ferroptosis-associated diseases. This mini-review aims to provide insights into vitamins closely linked to the regulation of ferroptosis from the perspective of cellular reactive oxygen species biology. The mainstream reprogramming mechanisms of ferroptosis are overviewed, focusing on unique biological processes of iron metabolism, lipid metabolism, and amino acid metabolism. Moreover, recent breakthroughs in therapeutic interventions targeting ferroptosis via fully utilizing vitamin-based pharmacological tools were overviewed, covering vitamins (B, C, E, and K). Finally, mechanism insight related to vitamin-associated nutrient signaling was provided, highlighting the pharmacological benefits of metabolically reprogramming ferroptosis-associated diseases.
Collapse
Affiliation(s)
- Junjie Wu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yanting Shi
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Man Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Min Chen
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Shuying Ji
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xingxing Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Rui Xia
- School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Xiaohua Zheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
7
|
Hammad G, Magdy M, Aboushousha T, Abdelraouf A, Mamdouh S. HEPPAR1 and PIWIL2 as Panel Markers for Hepatocellular Carcinoma. Asian Pac J Cancer Prev 2024; 25:2123-2131. [PMID: 38918675 PMCID: PMC11382836 DOI: 10.31557/apjcp.2024.25.6.2123] [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: 02/05/2024] [Indexed: 06/27/2024] Open
Abstract
OBJECTIVE The aim of this study was to evaluate the expression profiles of PIWI-like protein- 2 (PIWIL2), and HepPar1 and their immunohistochemical (IHC) characteristics in Hepatocellular Carcinoma (HCC), and determine their correlation with clinicopathological parameters of this type of cancer to determine their diagnostic value in combination. METHODS Seventy-five patients with HCC were assessed for the expression of PIWIL2 in serum and tissue using real-time polymerase chain reaction (RT-PCR) and IHC was performed for PIWIL2 and HepPar1 was performed on all patients. RESULTS A statistically significantly higher level of PIWIL2 was found in HCC compared to controls (p≤0.001). Both HepPar1 and PIWIL2 were detected in 84% of HCC cases, the diagnostic and prognostic factors for PIWIL2 were found to be significant in liver tumour tissue samples and non-tumorous sections p<0.001, and the same was observed for serum samples and results of healthy serum controls (p<0.001) when compared to AFP. CONCLUSION Our results affirm the hypothesis that reactivation of PIWI expression in various caner types is crucial for cancer development, and that a possible panel maybe used for these markers HCC diagnosis.
Collapse
Affiliation(s)
- Gehan Hammad
- Faculty of Biotechnology, October University for Modern Sciences & Arts (MSA), Giza, Egypt
| | - Mona Magdy
- Department of Pathology, Theodor Bilharz Research Institute, (TBRI), Giza, Egypt
| | - Tarek Aboushousha
- Department of Pathology, Theodor Bilharz Research Institute, (TBRI), Giza, Egypt
| | - Amr Abdelraouf
- Department of Hepatobiliopancreatic Surgery, National Hepatology and Tropical Medicine Research Institute, (NHTMRI), Cairo, Egypt
| | - Samah Mamdouh
- Department of Biochemistry and Molecular Biology, Theodor Bilharz Research Institute, Giza, Egypt
| |
Collapse
|
8
|
Zhang C, Wang W, Wu B. Molecular mechanism of WWP1-mediated ubiquitination modification affecting proliferation and invasion/migration of liver cancer cells. Kaohsiung J Med Sci 2024; 40:255-268. [PMID: 37997542 DOI: 10.1002/kjm2.12786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/16/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
Liver cancer is the most prevalent fatal malignancy across the globe. The present study aims to explore the molecular mechanism of E3 ligase WWP1 in liver cancer cell proliferation and invasion/migration. RT-qPCR and Western blot were performed to detect WWP1, KLF14, and VEPH1 expressions in liver cancer cell lines. Furthermore, WWP1 expression was silenced in cells, followed by the detection of cell viability, proliferation, and invasion/migration by CCK-8, colony formation, and Transwell assays, respectively. ChIP was used to analyze the binding relationship between WWP1 and KLF14. We measured the KLF14 ubiquitination level and KLF14 enrichment on the VEPH1 promoter after MG132 treatment. Dual-luciferase reporter assay was used to validate the binding relationship between KLF14 and VEPH1. Consequently, WWP1 was highly expressed in liver cancer cells; WWP1 silencing reduced the proliferation and invasion/migration of liver cancer cells. Mechanistically, WWP1 promoted KLF14 ubiquitination degradation; KLF14 was enriched on the VEPH1 promoter to promote its transcription and protein expression. Inhibiting KLF14 or VEPH1 partially minimized the inhibitory effect of WWP1 silencing on liver cancer cell proliferation and invasion/migration. In summary, WWP1 degrades KLF14 through ubiquitination, hence repressing VEPH1 expression and accelerating proliferation and invasion/migration of liver cancer cells.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Hepatobiliary Surgery, Wuhan No 1 Hospital, Wuhan, Hubei, China
| | - Wei Wang
- Department of Hepatobiliary Surgery, Wuhan No 1 Hospital, Wuhan, Hubei, China
| | - Biao Wu
- Department of Gastrointestinal surgery, Wuhan No 1 Hospital, Wuhan, Hubei, China
| |
Collapse
|
9
|
Mo JQ, Zhang SY, Li Q, Chen MX, Zheng YQ, Xie X, Zhang R, Wang SS. Immunomodulation of cuproptosis and ferroptosis in liver cancer. Cancer Cell Int 2024; 24:22. [PMID: 38200525 PMCID: PMC10777659 DOI: 10.1186/s12935-023-03207-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024] Open
Abstract
According to statistics, the incidence of liver cancer is increasing yearly, and effective treatment of liver cancer is imminent. For early liver cancer, resection surgery is currently the most effective treatment. However, resection does not treat the disease in advanced patients, so finding a method with a better prognosis is necessary. In recent years, ferroptosis and cuproptosis have been gradually defined, and related studies have proved that they show excellent results in the therapy of liver cancer. Cuproptosis is a new form of cell death, and the use of cuproptosis combined with ferroptosis to inhibit the production of hepatocellular carcinoma cells has good development prospects and is worthy of in-depth discussion by researchers. In this review, we summarize the research progress on cuproptosis combined with ferroptosis in treating liver cancer, analyze the value of cuproptosis and ferroptosis in the immune of liver cancer, and propose potential pathways in oncotherapy with the combination of cuproptosis and ferroptosis, which can provide background knowledge for subsequent related research.
Collapse
Affiliation(s)
- Jia-Qian Mo
- School of Life Sciences and Biopharmaceutics, Guang Dong Pharmaceutical University, Guangzhou, 51006, China
| | - Shen-Yan Zhang
- School of Life Sciences and Biopharmaceutics, Guang Dong Pharmaceutical University, Guangzhou, 51006, China
| | - Qiang Li
- School of Life Sciences and Biopharmaceutics, Guang Dong Pharmaceutical University, Guangzhou, 51006, China
| | - Mo-Xian Chen
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China and College of Life Sciences, Nanjing Forestry University, Nanjing, 210037, China
| | - Yue-Qing Zheng
- Guang Zhou Zengcheng District Centre for Disease Control and Prevention, Guang Dong, 511300, China
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Rongxin Zhang
- School of Life Sciences and Biopharmaceutics, Guang Dong Pharmaceutical University, Guangzhou, 51006, China.
| | - Shan-Shan Wang
- School of Life Sciences and Biopharmaceutics, Guang Dong Pharmaceutical University, Guangzhou, 51006, China.
| |
Collapse
|