1
|
Hai L, Bai XY, Luo X, Liu SW, Ma ZM, Ma LN, Ding XC. Prognostic modeling of hepatocellular carcinoma based on T-cell proliferation regulators: a bioinformatics approach. Front Immunol 2024; 15:1444091. [PMID: 39445019 PMCID: PMC11496079 DOI: 10.3389/fimmu.2024.1444091] [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: 06/05/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Background The prognostic value and immune significance of T-cell proliferation regulators (TCRs) in hepatocellular carcinoma (HCC) have not been previously reported. This study aimed to develop a new prognostic model based on TCRs in patients with HCC. Method This study used The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) and International Cancer Genome Consortium-Liver Cancer-Riken, Japan (ICGC-LIRI-JP) datasets along with TCRs. Differentially expressed TCRs (DE-TCRs) were identified by intersecting TCRs and differentially expressed genes between HCC and non-cancerous samples. Prognostic genes were determined using Cox regression analysis and were used to construct a risk model for HCC. Kaplan-Meier survival analysis was performed to assess the difference in survival between high-risk and low-risk groups. Receiver operating characteristic curve was used to assess the validity of risk model, as well as for testing in the ICGC-LIRI-JP dataset. Additionally, independent prognostic factors were identified using multivariate Cox regression analysis and proportional hazards assumption, and they were used to construct a nomogram model. TCGA-LIHC dataset was subjected to tumor microenvironment analysis, drug sensitivity analysis, gene set variation analysis, and immune correlation analysis. The prognostic genes were analyzed using consensus clustering analysis, mutation analysis, copy number variation analysis, gene set enrichment analysis, and molecular prediction analysis. Results Among the 18 DE-TCRs, six genes (DCLRE1B, RAN, HOMER1, ADA, CDK1, and IL1RN) could predict the prognosis of HCC. A risk model that can accurately predict HCC prognosis was established based on these genes. An efficient nomogram model was also developed using clinical traits and risk scores. Immune-related analyses revealed that 39 immune checkpoints exhibited differential expression between the high-risk and low-risk groups. The rate of immunotherapy response was low in patients belonging to the high-risk group. Patients with HCC were further divided into cluster 1 and cluster 2 based on prognostic genes. Mutation analysis revealed that HOMER1 and CDK1 harbored missense mutations. DCLRE1B exhibited an increased copy number, whereas RAN exhibited a decreased copy number. The prognostic genes were significantly enriched in tryptophan metabolism pathways. Conclusions This bioinformatics analysis identified six TCR genes associated with HCC prognosis that can serve as diagnostic markers and therapeutic targets for HCC.
Collapse
Affiliation(s)
- Long Hai
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiao-Yang Bai
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xia Luo
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Infectious Disease Clinical Research Center of Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shuai-Wei Liu
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Infectious Disease Clinical Research Center of Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zi-Min Ma
- Weiluo Microbial Pathogens Monitoring Technology Co., Ltd. of Beijing, Beijing, China
| | - Li-Na Ma
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Infectious Disease Clinical Research Center of Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiang-Chun Ding
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Infectious Disease Clinical Research Center of Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Tropical Disease & Infectious Disease, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
2
|
Zhang Q, Yang G, Chang R, Wang F, Han T, Tian J, Wang W. Time series analysis combined with transcriptome sequencing to explore characteristic genes and potential molecular mechanisms associated with ultrasound-guided microwave ablation of glioma. Int J Hyperthermia 2024; 41:2406889. [PMID: 39317933 DOI: 10.1080/02656736.2024.2406889] [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: 07/13/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024] Open
Abstract
OBJECTIVE This study aimed to explore marker genes and their potential molecular mechanisms involved in US-guided MWA for glioma in mice. METHOD The differentially expressed genes (DEGs1 and DEGs2) and lncRNAs (DELs1 and DELs2) were obtained between Non (glioma tissues without MWA) and T0 groups (0h after MWA), as well as between Non and T24 groups (24h after MWA). The down-regulation cluster genes (CONDOWNDEGs) and upregulation cluster genes (CONUPDEGs) were identified by time series analysis. Candidate genes were obtained by overlapping CONDOWNDEGs with downregulation DEGs (DOWNDEGs)1 and DOWNDEGs2, as well as CONUPDEGs with up-regulation DEGs (UPDEGs)1 and UPDEGs2. The expressions of immune checkpoints and inflammatory factors, gene set enrichment analysis (GSEA), and protein subcellular localization were performed. The eXpression2Kinases (X2K), GeneMANIA, transcription factor (TF), and competing endogenous (ce) RNA regulatory networks were conducted. The expression of marker genes was validated by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS Five marker genes (IL32, VCAM1, IL34, NFKB1 and CXCL13) were identified, which were connected with immune-related functions. Two immune checkpoints (CD96 and TIGIT) and six inflammatory factors played key roles in US-guided MWA for glioma. ceRNA regulatory networks revealed that miR-625-5p, miR-625-3p, miR-31-5p and miR-671-5p were associated with target genes. qRT-PCR indicated both IL32, VCAM1, and NFKB1 were potential markers under US-guided MWA-related time series analysis. CONCLUSION The use of US-guided MWA might be a practical method for influencing the function of target genes, regulating time frames to decrease inflammation, and stimulating immune responses in glioma therapy.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Cardiovascular and Cerebrovascular Disease Hospital, Yinchuan, China
| | - Guangfei Yang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ruijiao Chang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Fuxia Wang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Cardiovascular and Cerebrovascular Disease Hospital, Yinchuan, China
| | - Tao Han
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jin Tian
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wen Wang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Cardiovascular and Cerebrovascular Disease Hospital, Yinchuan, China
| |
Collapse
|
3
|
Na X, Li L, Liu D, He J, Zhang L, Zhou Y. Natural products targeting ferroptosis pathways in cancer therapy (Review). Oncol Rep 2024; 52:123. [PMID: 39054952 PMCID: PMC11292301 DOI: 10.3892/or.2024.8782] [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: 04/14/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Ferroptosis inducers (FIN) have a key role in cancer therapy and provide novel and innovative treatment strategies. Although many researchers have performed FIN screening of synthetic compounds, studies on the identification of FIN from natural products are limited, particularly in the field of drug development and combination therapy. In this review, this gap was addressed by comprehensively summarizing recent studies on ferroptosis. The causes of ferroptosis were categorized into driving and defensive factors, elucidating key pathways and targets. Next, through summarizing research on natural products that induce ferroptosis, the study elaborated in detail on the natural products that have FIN functions. Their discovery and development were also described and insight for clinical drug development was provided. In addition, the mechanisms of action were analyzed and potential combination therapies, resistance reversal and structural enhancements were presented. By highlighting the potential of natural products in inducing ferroptosis for cancer treatment, this review may serve as a reference for utilizing these compounds against cancer. It not only showed the significance of natural products but may also promote further investigation into their therapeutic effects, thus encouraging research in this field.
Collapse
Affiliation(s)
- Xin Na
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Lin Li
- Yunnan Cancer Hospital (Third Affiliated Hospital of Kunming Medical University), Kunming, Yunnan 650118, P.R. China
| | - Dongmei Liu
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Jiaqi He
- The First Clinical Medical College of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Ling Zhang
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yiping Zhou
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
4
|
Wang H, Xie M, Zhao Y, Zhang Y. Establishment of a prognostic risk model for prostate cancer based on Gleason grading and cuprotosis related genes. J Cancer Res Clin Oncol 2024; 150:376. [PMID: 39085482 PMCID: PMC11291559 DOI: 10.1007/s00432-024-05899-9] [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: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION Prostate cancer (PCa) is common in aging males, diagnosed via the Gleason grading system. The study explores the unexamined prognostic value of cuprotosis, a distinct cell death type, alongside Gleason grades in PCa. METHODS We explored Cuprotosis-related genes (CRGs) in prostate cancer (PCa), using NMF on TCGA-PRAD data for patient classification and WGCNA to link genes with Gleason scores and prognosis. A risk model was crafted via LASSO Cox regression. STX3 knockdown in PC-3 cells, analyzed for effects on cell behaviors and tumor growth in mice, highlighted its potential therapeutic impact. RESULTS We identified five genes crucial for a prognostic risk model, with higher risk scores indicating worse prognosis. Survival analysis and ROC curves confirmed the model's predictive accuracy in TCGA-PRAD and GSE70769 datasets. STX3 was a key adverse prognostic factor, with its knockdown significantly reducing mRNA and protein levels, impairing PC-3 cell functions. In vivo, STX3 knockdown in PC-3 cells led to significantly smaller tumors in nude mice, underscoring its potential therapeutic value. CONCLUSION Our prognostic model, using five genes linked to Gleason scores, effectively predicts prostate cancer outcomes, offering a novel treatment strategy angle.
Collapse
Affiliation(s)
- Haicheng Wang
- Department of Urology, Hebei Medical University, Shijiazhuang, China
- Department of Urology, Qinhuangdao First Hospital, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, China
| | - Meiyi Xie
- Department of Urology, Qinhuangdao First Hospital, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, China
| | - Yuming Zhao
- Department of Urology, Qinhuangdao First Hospital, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, China
| | - Yong Zhang
- Department of Urology, Hebei Medical University, Shijiazhuang, China.
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Wen Z, Zhang Y, Gao B, Chen X. Baicalin induces ferroptosis in oral squamous cell carcinoma by suppressing the activity of FTH1. J Gene Med 2024; 26:e3669. [PMID: 38380717 DOI: 10.1002/jgm.3669] [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: 12/06/2023] [Revised: 01/02/2024] [Accepted: 01/14/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND This study investigated the role of the ferroptosis-related gene FTH1 in oral squamous cell carcinoma (OSCC) and evaluated the therapeutic potential of baicalin in OSCC cell treatment. METHODS A prognostic model was established by bioinformatic analysis, consisting of 12 ferroptosis related genes (FRGs), and FTH1 was selected as the most significantly up-regulated FRGs. The clinical correlation of FTH1 in OSCC samples was evaluated by both immunohistochemical and bioinformatic characterizations. The effects of FTH1 on migration, invasion, epithelial-mesenchymal transition (EMT) and proliferation were determined by wound healing assays, transwell assays, western blotting and 5'-ethynl 2'-deoxyuridine proliferation assays, respectively. The effects of FTH1 on ferroptosis were tested via ferroptosis markers and Mito Tracker staining. In addition, the therapeutic effects of baicalin on OSCC cells were confirmed using EMT, migration, invasion, proliferation and ferroptosis assays. RESULTS The 12 FRGs were predictive of the prognosis for OSCC patients, and FTH1 expression was identified as significantly up-regulated in OSCC samples, which was highly associated with survival, immune cell infiltration and drug sensitivity. Moreover, knocking down FTH1 inhibited cell proliferation, EMT and invasive phenotypes, but induced ferroptosis in OSCC cells (Cal27 and SCC25). Furthermore, baicalin directly suppressed expression of FTH1 in OSCC cells, and effectively promoted ferroptosis and inhibited the proliferation as well as EMT by directly targeting FTH1. CONCLUSIONS This study has demonstrated that FTH1 is a therapeutic target for OSCC treatment, and has provided evidence that baicalin offers a promising alternative for OSCC treatment.
Collapse
Affiliation(s)
- Zhihao Wen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxiao Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Bo Gao
- Kunming Medical University, Kunming, China
| | - Xin Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Zhu J, Shen P, Xu Y, Zhang X, Chen Q, Gu K, Ji S, Yang B, Zhao Y. Ferroptosis: a new mechanism of traditional Chinese medicine for cancer treatment. Front Pharmacol 2024; 15:1290120. [PMID: 38292937 PMCID: PMC10824936 DOI: 10.3389/fphar.2024.1290120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
Ferroptosis, distinct from apoptosis, is a novel cellular death pathway characterized by the build-up of lipid peroxidation and reactive oxygen species (ROS) derived from lipids within cells. Recent studies demonstrated the efficacy of ferroptosis inducers in targeting malignant cells, thereby establishing a promising avenue for combating cancer. Traditional Chinese medicine (TCM) has a long history of use and is widely used in cancer treatment. TCM takes a holistic approach, viewing the patient as a system and utilizing herbal formulas to address complex diseases such as cancer. Recent TCM studies have elucidated the molecular mechanisms underlying ferroptosis induction during cancer treatment. These studies have identified numerous plant metabolites and derivatives that target multiple pathways and molecular targets. TCM can induce ferroptosis in tumor cells through various regulatory mechanisms, such as amino acid, iron, and lipid metabolism pathways, which may provide novel therapeutic strategies for apoptosis-resistant cancer treatment. TCM also influence anticancer immunotherapy via ferroptosis. This review comprehensively elucidates the molecular mechanisms underlying ferroptosis, highlights the pivotal regulatory genes involved in orchestrating this process, evaluates the advancements made in TCM research pertaining to ferroptosis, and provides theoretical insights into the induction of ferroptosis in tumors using botanical drugs.
Collapse
Affiliation(s)
- Jiahao Zhu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Clinical Cancer Center, Wuxi, Jiangsu, China
| | - Peipei Shen
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Clinical Cancer Center, Wuxi, Jiangsu, China
| | - Yu Xu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Clinical Cancer Center, Wuxi, Jiangsu, China
| | - Xiaojun Zhang
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Clinical Cancer Center, Wuxi, Jiangsu, China
| | - Qingqing Chen
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Ke Gu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Clinical Cancer Center, Wuxi, Jiangsu, China
| | - Shengjun Ji
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Bo Yang
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Clinical Cancer Center, Wuxi, Jiangsu, China
| | - Yutian Zhao
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Clinical Cancer Center, Wuxi, Jiangsu, China
| |
Collapse
|
7
|
Ke S, Pan Q, Wang C, Su Z, Li M, Liu X. NKD2 Trigger NF-κB Signaling Pathway and Facilitates Thyroid Cancer Cell Proliferation. Mol Biotechnol 2023; 65:1846-1856. [PMID: 36820951 PMCID: PMC10518272 DOI: 10.1007/s12033-023-00665-7] [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: 10/11/2022] [Accepted: 01/12/2023] [Indexed: 02/24/2023]
Abstract
NKD inhibitor of WNT signaling pathway 2 (NKD2) is an emerging player in cancer onset and progression. Here, it was confirmed that THCA patients have robustly expressed NKD2, which was linked to an advanced pathologic stage. The prognosis was worse for those with high NKD2 levels. Functionally, ectopically produced NKD2 promotes THCA cell proliferation, whereas NKD2 knockdown impairs the ability of THCA cells to proliferate. Mechanically, ectopically expressed NKD2 activated NF-κB transcriptional activity, whereas NKD2-deficient THCA cells showed lower NF-κB transcriptional activity. As a result, NKD2 activates the NF-κB signaling pathway, encouraging the growth of THCA cells.
Collapse
Affiliation(s)
- Shaoying Ke
- Department of Throid Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Qunxiong Pan
- Department of Throid Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Congren Wang
- Department of Throid Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Zijian Su
- Department of Throid Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Mingzhu Li
- Department of Throid Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Xiaoyu Liu
- Department of Throid Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000, Fujian, China.
| |
Collapse
|
8
|
Zhang P, Liu W, Wang Y. The mechanisms of tanshinone in the treatment of tumors. Front Pharmacol 2023; 14:1282203. [PMID: 37964867 PMCID: PMC10642231 DOI: 10.3389/fphar.2023.1282203] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Tanshinone is a lipophilic compound that is present in traditional Chinese medicine and is derived from the roots of Salvia miltiorrhiza (Danshen). It has been proven to be highly effective in combating tumors in various parts of the body, including liver carcinoma, gastric cancer, ovarian cancer, cervix carcinoma, breast cancer, colon cancer, and prostate cancer. Tanshinone can efficiently prevent the reproduction of cancerous cells, induce cell death, and inhibit the spread of cancerous cells, which are mainly involved in the PI3K/Akt signaling pathway, NF-κB pathway, Bcl-2 family, Caspase cascades, MicroRNA, MAPK signaling pathway, p21, STAT3 pathway, miR30b-P53-PTPN11/SHP2 axis, β-catenin, and Skp2. However, the properties and mechanisms of tanshinone's anti-tumor effects remain unclear currently. Thus, this study aims to review the research progress on tumor prevention and mechanisms of tanshinone to gain new perspectives for further development and clinical application of tanshinone.
Collapse
Affiliation(s)
- Pengyu Zhang
- The Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wendi Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Wang
- Department of Histology and Embryology, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
9
|
Huang Y, Li X, Zhang Z, Xiong L, Wang Y, Wen Y. Photodynamic Therapy Combined with Ferroptosis Is a Synergistic Antitumor Therapy Strategy. Cancers (Basel) 2023; 15:5043. [PMID: 37894410 PMCID: PMC10604985 DOI: 10.3390/cancers15205043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/20/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Ferroptosis is a programmed death mode that regulates redox homeostasis in cells, and recent studies suggest that it is a promising mode of tumor cell death. Ferroptosis is regulated by iron metabolism, lipid metabolism, and intracellular reducing substances, which is the mechanism basis of its combination with photodynamic therapy (PDT). PDT generates reactive oxygen species (ROS) and 1O2 through type I and type II photochemical reactions, and subsequently induces ferroptosis through the Fenton reaction and the peroxidation of cell membrane lipids. PDT kills tumor cells by generating excessive cytotoxic ROS. Due to the limited laser depth and photosensitizer enrichment, the systemic treatment effect of PDT is not good. Combining PDT with ferroptosis can compensate for these shortcomings. Nanoparticles constructed by photosensitizers and ferroptosis agonists are widely used in the field of combination therapy, and their targeting and biological safety can be improved through modification. These nanoparticles not only directly kill tumor cells but also further exert the synergistic effect of PDT and ferroptosis by activating antitumor immunity, improving the hypoxia microenvironment, and inhibiting the tumor angiogenesis. Ferroptosis-agonist-induced chemotherapy and PDT-induced ablation also have good clinical application prospects. In this review, we summarize the current research progress on PDT and ferroptosis and how PDT and ferroptosis promote each other.
Collapse
Affiliation(s)
- Yunpeng Huang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Xiaoyu Li
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China;
| | - Zijian Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Li Xiong
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Yongxiang Wang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Yu Wen
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| |
Collapse
|
10
|
Huang L, Chen G, He J, Wang P. ZC3H13 reduced DUOX1-mediated ferroptosis in laryngeal squamous cell carcinoma cells through m6A-dependent modification. Tissue Cell 2023; 84:102187. [PMID: 37536262 DOI: 10.1016/j.tice.2023.102187] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is the second most common head and neck cancer. To identify the link between ferroptosis and LSCC, we targeted the dual oxidase 1 (DUOX1) gene. This study aimed to reveal the intrinsic mechanism by which the DUOX1-zinc-finger CCCH domain-containing protein 13 (ZC3H13) ferroptosis axis affected the LSCC process. GEPIA was used to investigate the expression of DUOX1 in LSCC, and the expression levels of DUOX1 and ZC3H13 were manipulated by overexpression and RNA interference. MTT assay was used to detect cell proliferation. Chromatin immunoprecipitation (CHIP) detected the binding of DUOX1 and ZC3H13, and ROS assessment and intracellular Fe2+ content determination were performed to examine the ferroptosis. MeRIP was used to analyze the m6A methylation of DUOX1. Ferroptosis-related proteins were detected by qRT-PCR. DUOX1 was found to be poorly expressed in LSCC cells, low DUOX1 level promoted LSCC cell proliferation, and low ZC3H13 level decreased LSCC cell proliferation. Besides, there was an interaction between DUOX1 and ZC3H13. DUOX1 could inhibit the expression levels of ferroptosis-related genes GPX4 and F1H1 in LSCC cells DUOX1 inhibited the expression levels of ROS and ferroptosis-related genes GPX4 and F1H1 and increased intracellular iron content in LSCC cells, but ZC3H13 reversed this phenomenon by inhibiting DUOX1 gene through m6A methylation modification. ZC3H13 reduced DUOX1-mediated ferroptosis in LSCC cells through m6A-dependent modification. The regulatory pathway of DUOX1 and ferroptosis are potential targets for designing diagnostic and combination therapeutic strategies for LSCC patients.
Collapse
Affiliation(s)
- Lili Huang
- Department of Otorhinolaryngology, The First People's Hospital of Fuyang Hangzhou, 311400 Hangzhou, Zhejiang Province, China
| | - Guangli Chen
- Department of Otorhinolaryngology, The First People's Hospital of Fuyang Hangzhou, 311400 Hangzhou, Zhejiang Province, China
| | - Jing He
- Department of Otorhinolaryngology, The First People's Hospital of Fuyang Hangzhou, 311400 Hangzhou, Zhejiang Province, China
| | - Pu Wang
- Department of Otorhinolaryngology, The First People's Hospital of Fuyang Hangzhou, 311400 Hangzhou, Zhejiang Province, China.
| |
Collapse
|
11
|
Belvin BR, Lewis JP. Ferroportin depletes iron needed for cell cycle progression in head and neck squamous cell carcinoma. Front Oncol 2023; 12:1025434. [PMID: 36698390 PMCID: PMC9868905 DOI: 10.3389/fonc.2022.1025434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/16/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Ferroportin (FPN), the only identified eukaryotic iron efflux channel, plays an important role in iron homeostasis and is downregulated in many cancers. To determine if iron related pathways are important for Head and Neck Squamous Cell Carcinoma (HNSCC) progression and proliferation, we utilize a model of FPN over-expression to simulate iron depletion and probe associated molecular pathways. Methods The state of iron related proteins and ferroptosis sensitivity was assessed in a panel of metastatic HNSCC cell lines. Stable, inducible expression of FPN was confirmed in the metastatic HNSCC lines HN12 and JHU-022 as well as the non-transformed normal oral keratinocyte (NOK) cell line and the effect of FPN mediated iron depletion was assessed in these cell lines. Results HNSCC cells are sensitive to iron chelation and ferroptosis, but the non-transformed NOK cell line is not. We found that FPN expression inhibits HNSCC cell proliferation and colony formation but NOK cells are unaffected. Inhibition of cell proliferation is rescued by the addition of hepcidin. Decreases in proliferation are due to the disruption of iron homeostasis via loss of labile iron caused by elevated FPN levels. This in turn protects HNSCC cells from ferroptotic cell death. Expression of FPN induces DNA damage, activates p21, and reduces levels of cyclin proteins thereby inhibiting cell cycle progression of HNSCC cells, arresting cells in the S-phase. Induction of FPN severely inhibits Edu incorporation and increased β-galactosidase activity, indicating cells have entered senescence. Finally, in an oral orthotopic mouse xenograft model, FPN induction yields a significant decrease in tumor growth. Conclusions Our results indicate that iron plays a role in HNSCC cell proliferation and growth and is important for cell cycle progression. Iron based interventional strategies such as ferroptosis or iron chelation may have potential therapeutic benefits in advanced HNSCC.
Collapse
Affiliation(s)
- Benjamin Ross Belvin
- Philips Institute for Oral Health Research, School of Dentistry, Richmond, VA, United States
| | - Janina P. Lewis
- Philips Institute for Oral Health Research, School of Dentistry, Richmond, VA, United States,Department of Biochemistry and Molecular Biology, School of Medicine, Richmond, VA, United States,Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States,*Correspondence: Janina P. Lewis,
| |
Collapse
|