1
|
Wu YF, Jiang XH, Qian DT. Establishment and validation of a novel risk model based on PANoptosis-related genes to predict prognosis in head and neck squamous cell carcinoma. Medicine (Baltimore) 2025; 104:e42299. [PMID: 40324259 PMCID: PMC12055095 DOI: 10.1097/md.0000000000042299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/27/2024] [Accepted: 04/11/2025] [Indexed: 05/07/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSC) is a common cancer worldwide with poor prognosis. Current treatment methods have limited effect on improving the prognosis of patients with HNSC. Differentially expressed PANoptosis-related genes in HNSC were identified from the TCGA using limma and WGCNA. A prognostic model was established using univariate and multivariate Cox regression analyses and machine learning, and its performance was evaluated using Kaplan-Meier and receiver operating characteristic curves. SNP data was analyzed using maftools package. Immune analysis was performed using IOBR package and TIDE website. The scRNA data was analyzed using Seurat and cellchat package. The expression of hub genes was validated in vitro. The prognostic model comprising 5 hub PANoptosis-related genes (AIFM1, AKT3, CDKN2A, EGFR, IL1RAP) accurately predicted patient outcomes, with the high-risk group exhibiting poorer survival. mRNA expression levels of all 5 hub genes were elevated in the tumor cells, but only AIFM1, AKT3 and IL1RAP's protein expression were higher in tumor tissues. Additionally, high expression of AIFM1, AKT3, EGFR, IL1RAP and low expression of CDKN2A indicated poor prognosis of HNSC patients. The decreasing levels of CD4 T cells, CD8 T cells and M1 macrophages were observed in high-risk groups. There was a significant difference of 5-fluorouracil in low and high-risk groups. scRNA analysis exhibited that TNF pathway was important in the interaction between macrophages and T cells. We identified 5 hub genes and constructed a great risk model for the prognosis of HNSC. The immune cells may influence the HNSC malignant through TNF signal pathway.
Collapse
Affiliation(s)
- Yi-Fen Wu
- Department of Stomatology, People’s Hospital of Kecheng District, Quzhou City, Zhejiang, China
| | - Xiao-Hui Jiang
- Department of Stomatology, People’s Hospital of Kecheng District, Quzhou City, Zhejiang, China
| | - Dan-Ting Qian
- Department of Stomatology, People’s Hospital of Kecheng District, Quzhou City, Zhejiang, China
| |
Collapse
|
2
|
Li W, Chen Y, Zhang Y, Wen W, Lu Y. Comprehensive analysis of the relationship between RNA modification writers and immune microenvironment in head and neck squamous cell carcinoma. BMC Immunol 2024; 25:76. [PMID: 39533178 PMCID: PMC11558979 DOI: 10.1186/s12865-024-00667-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. Four types of RNA modification writers (m6A, m1A, A-I editing, and APA) are widely involved in tumorigenesis and the TME. We aimed to comprehensively explore the role of the four RNA modification writers in the progression and immune microenvironment of HNSCC. MATERIALS AND METHODS We first obtained transcription profile data and transcriptional variation of the four types of RNA modification writers from The Cancer Genome Atlas (TCGA) database. HNSCC patients in TCGA dataset were divided into different clusters based on the four types of RNA modification writers. Univariate Cox and Least absolute shrinkage and selection operator (LASSO) analyses were performed to conduct a Writer-score scoring system, which was successfully verified in the GSE65858 dataset and our clinical sample dataset. Finally, we evaluated the relationship between different RNA modification clusters (Writer-score) and immunological characteristics of HNSCC. RESULTS Two different RNA modification clusters (A and B) were obtained. These RNA modification clusters (Writer-score) were strongly associated with immunological characteristics (immunomodulators, cancer immunity cycles, infiltrating immune cells (TIICs), inhibitory immune checkpoints, and T cell inflamed score (TIS)) of HNSCC. CONCLUSIONS This study identified two different RNA modification clusters and explored the potential relationship between RNA modification clusters (Writer-score) and immunological characteristics, offering a new theoretical basis for precision immunotherapy in patients with HNSCC.
Collapse
Affiliation(s)
- Wei Li
- The First Clinical College of China Medical University, Shenyang, China
| | - Ying Chen
- Department of Ultrasound, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, China
| | - Yao Zhang
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wen Wen
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yingying Lu
- The First Clinical College of Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
3
|
Gong Y, Gong D, Liu S, Gong X, Xiong J, Zhang J, Jiang L, Liu J, Zhu L, Luo H, Xu K, Yang X, Li B. Deciphering the role of NcRNAs in Pancreatic Cancer immune evasion and drug resistance: a new perspective for targeted therapy. Front Immunol 2024; 15:1480572. [PMID: 39555076 PMCID: PMC11563824 DOI: 10.3389/fimmu.2024.1480572] [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: 08/14/2024] [Accepted: 10/09/2024] [Indexed: 11/19/2024] Open
Abstract
Pancreatic cancer (PC) is a very aggressive digestive system tumor, known for its high mortality rate, low cure rate, low survival rate and poor prognosis. In particular, pancreatic ductal adenocarcinoma (PADC), which accounts for more than 90% of PC cases, has an overall 5-year survival rate of only 5%, which is an extremely critical situation. Early detection and effective treatment of PC is extremely difficult, which leads many patients to despair. In the current medical context, targeted therapy, as an important strategy for cancer treatment, is expected. However, the problems of immune escape and drug resistance in PC have become two major obstacles that are difficult to be overcome by targeted therapy. How to break through these two difficulties has become a key issue to be solved in the field of PC therapy. In recent years, non-coding RNAs (ncRNAs) have continued to heat up in the field of cancer research. NcRNAs play a pivotal role in gene regulation, cell differentiation, development, and disease processes, and their important roles in the genesis, development, and therapeutic response of PC have been gradually revealed. More importantly, ncRNAs have many advantages as therapeutic targets, such as high specificity and low side effects, making them a new favorite in the field of PC therapy. Therefore, the aim of this paper is to provide new ideas and methods for the targeted therapy of PC by reviewing the mechanism of action of four major ncRNAs (circRNAs, lncRNAs, miRNAs, siRNAs) in both immune escape and drug resistance of PC. It is expected that an effective way to overcome immune escape and drug resistance can be found through in-depth study of ncRNA, bringing a ray of hope to PC patients.
Collapse
Affiliation(s)
- Yu Gong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Desheng Gong
- Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- General Surgery Department, The TCM Hospital of Longquanyi, Chengdu, China
| | - Sinian Liu
- Department of Pathology, Xichong People’s Hospital, Nanchong, China
| | - Xiangjin Gong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Jingwen Xiong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Jinghan Zhang
- Department of Anesthesia, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jie Liu
- Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Lin Zhu
- Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Huiyang Luo
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Xiaoli Yang
- Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Zhang X, Hong B, Li H, Sun Z, Zhao J, Li M, Wei D, Wang Y, Zhang N. Disulfidptosis and ferroptosis related genes define the immune microenvironment and NUBPL serves as a potential biomarker for predicting prognosis and immunotherapy response in bladder cancer. Heliyon 2024; 10:e37638. [PMID: 39290277 PMCID: PMC11407088 DOI: 10.1016/j.heliyon.2024.e37638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/05/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
Background Ferroptosis and disulfidptosis are regulatory forms of cell death that play an important role in tumorigenesis and progression. However, few biomarkers about disulfidptosis and ferroptosis related genes (DFRGs) have been developed to predict the prognosis of bladder cancer (BC). Methods We conducted a bioinformatics analysis using public BC datasets to examine the prognostic significance of differentially expressed DFRGs. A Lasso regression was employed to create a prognostic prediction model from these DFRGs. Hub DFRGs that play a role in immunotherapy response and immunoregulation were pinpointed. Immunohistochemistry (IHC) experiment was performed to assess NUBPL and c-MYC expression in BC patients who underwent surgery or received immune checkpoint inhibitor (ICI) immunotherapy at Peking University Cancer Hospital. Results We constructed a valid model to predict the prognosis of BC based on DFRGs and performed relevant validation, the results demonstrated that the model was an independent prognostic factor for BC. Further analysis indicated that the model score, combined with the expression of various immune factors and tumor mutation burden (TMB), could predict the prognosis for BC. In addition, we also found that NUBPL was strongly associated with prognosis and response to ICI treatment, and NUBPL may influence BC malignant progression through the c-MYC pathway. Conclusions Our research findings highlight the satisfactory predictive value of DFRGs in the immune microenvironment and suggest that NUBPL may be a highly promising biomarker for predicting the prognosis and efficacy of ICI treatment in BC patients.
Collapse
Affiliation(s)
- Xuezhou Zhang
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Baoan Hong
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Hongwei Li
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, PR China
| | - Zhipeng Sun
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Jiahui Zhao
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Mingchuan Li
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Dechao Wei
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Yongxing Wang
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Ning Zhang
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| |
Collapse
|
5
|
Xu C, Ma H, Chen J, Li X, Wang Z, Hu B, Zhang N, Meng F. Prognostic, immunity, stemness, and anticancer drug sensitivity characterization of pyroptosis related genes in non-small cell lung cancer. Thorac Cancer 2024; 15:215-226. [PMID: 38115677 PMCID: PMC10803221 DOI: 10.1111/1759-7714.15180] [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: 07/23/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Pyroptosis plays a pivotal role in the tumor immune microenvironment (TME) dynamics, particularly in non-small cell lung cancer (NSCLC). The aim of our study was to explore its effects on tumor progression, TME patterns, and the efficacy of therapeutic interventions in NSCLC. METHODS Our investigation encompassed a thorough analysis of pyroptosis-related genes (PRGs), integrating immunohistochemistry (IHC) data, TME characteristics, stemness indices, and anticancer drug sensitivities. We aimed to analyze mRNA expression profiles across various cancers, constructing benchmark datasets to assess the clinical significance of PRGs in NSCLC. This included evaluating their association with clinical responses and efficacy. Notably, both our and HPA IHC data demonstrated significantly elevated GSDMD-N protein levels in lung squamous cell carcinoma (LUSC) tissues. RESULTS The expression of PRGs differed significantly between tumor and normal tissues across various cancers, as validated by IHC data, and was correlated with prognosis (p < 0.05). Moreover, our investigation revealed significant differences (p < 0.05) in the expression of the PRGs among distinct TME subtypes categorized as C1 (wound healing), C3 (inflammatory), C2 (IFN-gamma dominant), C5 (immunological quiet), C4 (lymphocyte deficient), and C6 (TGF-beta dominant). Additionally, our research on anticancer drug sensitivity uncovered compelling connections between specific anticancer medications and the expression of PRGs, including GSDMD, ELANE, IL18, and CHMP4A (p < 0.05). CONCLUSION Our study provided valuable insights into the critical role of PRGs in TME modulation, tumor stemness, and anticancer drug sensitivity across diverse cancers. Our findings illuminate the intricate relationship between pyroptosis and the TME, offering new perspectives for enhancing NSCLC treatment and prognosis.
Collapse
Affiliation(s)
- Cong Xu
- Department of Thoracic SurgeryPeking University Shougang HospitalBeijingChina
| | - Hongming Ma
- Department of Respiratory and Critical CareEmergency General HospitalBeijingChina
| | | | - Xincheng Li
- Department of Thoracic SurgeryBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Zhina Wang
- Department of Respiratory and Critical CareEmergency General HospitalBeijingChina
| | - Bin Hu
- Department of Thoracic SurgeryBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Nan Zhang
- Department of Respiratory and Critical CareEmergency General HospitalBeijingChina
| | - Fanjie Meng
- Department of Thoracic SurgeryBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
6
|
He R, Zhang X, Wu Y, Weng Z, Li L. TTC7B is a new prognostic biomarker in head and neck squamous cell carcinoma linked to immune infiltration and ferroptosis. Cancer Med 2023; 12:22354-22369. [PMID: 37990988 PMCID: PMC10757123 DOI: 10.1002/cam4.6715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023] Open
Abstract
OBJECTIVE To investigate the expression of TTC7B and its prognostic significance, biological roles, and impact on the immune system in patients with head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS Clinical and genomic data were obtained from TCGA (The Cancer Genome Atlas), GEO (Gene Expression Omnibus), GEPIA2 (Gene Expression Profiling Interactive Analysis 2.0), and TIMER2.0 (Tumor Immune Estimation Resource 2.0) databases. R software was utilized to process the retrieved data. qPCR and immunohistochemical assays were performed to validate the findings obtained from the databases. RESULTS High expression of TTC7B was observed in HNSCC, and this heightened expression is significantly associated with reduced overall survival (OS) in patients, making it an independent risk factor impacting OS. TTC7B is correlated with focal adhesions and cell migration pathways based on functional enrichment analysis. CIBERSORT analysis and TIMER2.0 show a positive link between TTC7B and multiple immune cells, particularly macrophages. Pearson's analysis reveals a significant correlation between TTC7B and ferroptosis-related genes. CONCLUSION In all, TTC7B could serve as a promising prognostic indicator of HNSCC, and is closely associated with focal adhesions, immune infiltration, and ferroptosis.
Collapse
Affiliation(s)
- Rong He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Xun Zhang
- Guangyuan Hospital of Traditional Chinese MedicineGuangyuanChina
| | - Yongzhi Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Zhijie Weng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
7
|
Xu C, Xu H, Liu B. Head and neck squamous cell carcinoma-specific prognostic signature and drug sensitive subtypes based on programmed cell death-related genes. PeerJ 2023; 11:e16364. [PMID: 38025757 PMCID: PMC10668860 DOI: 10.7717/peerj.16364] [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: 05/08/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Background As a complex group of malignancies, head and neck squamous cell carcinoma (HNSC) is one of the leading causes of cancer mortality. This study aims to establish a reliable clinical classification and gene signature for HNSC prognostic prediction and precision treatments. Methods A consensus clustering analysis was performed to group HNSC patients in The Cancer Genome Atlas (TCGA) database based on genes linked to programmed cell death (PCD). Differentially expressed genes (DEGs) between subtypes were identified using the "limma" R package. The TCGA prognostic signature and PCD-related prognostic genes were found using a least absolute shrinkage and selection operator (LASSO) regression analysis and univariate Cox regression analysis. The robustness of the LASSO analysis was validated using datasets GSE65858 and GSE41613. A cell counting kit-8 (CCK-8) test, Western blot, and real-time reverse transcriptase-polymerase chain reaction (RT-qPCR) were used to evaluate the expression and viability of prognostic genes. Results Four molecular subtypes were identified in PCD-related genes. Subtype C4 had the best prognosis and the highest immune score, while subtype C1 exhibited the most unfavorable outcomes. Three hundred shared DEGs were identified among the four subtypes, and four prognostic genes (CTLA4, CAMK2N1, PLAU and CALML5) were used to construct a TCGA-HNSC prognostic model. High-risk patients manifested poorer prognosis, more inflammatory pathway enrichment, and lower immune cell infiltration. High-risk patients were more prone to immune escape and were more likely to be resistant to Cisplatin and 5-Fluorouracil. Prognosis prediction was validated in external datasets. The expression of CTLA4, CAMK2N1, PLAU and CALML5 was enhanced in CAL-27 and SCC-25 cell lines, and CALML5 inhibited CAL-27 and SCC-25 cell viability. Conclusion This study shares novel insights into HNSC classification and provides a reliable PCD-related prognostic signature for prognosis prediction and treatment for patients with HNSC.
Collapse
Affiliation(s)
- Chengbo Xu
- Department of Otolaryngology Head and Neck Surgery, Jinhua Wenrong Hospital, Jinhua, China
| | - Hongfang Xu
- Department of Otolaryngology Head and Neck Surgery, Jinhua Wenrong Hospital, Jinhua, China
| | - Baimei Liu
- Department of Otolaryngology Head and Neck Surgery, Yongkang First People’s Hospital, Yongkang, China
| |
Collapse
|
8
|
Li Y, Du Y, Zhou Y, Chen Q, Luo Z, Ren Y, Chen X, Chen G. Iron and copper: critical executioners of ferroptosis, cuproptosis and other forms of cell death. Cell Commun Signal 2023; 21:327. [PMID: 37974196 PMCID: PMC10652626 DOI: 10.1186/s12964-023-01267-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/11/2023] [Indexed: 11/19/2023] Open
Abstract
Regulated cell death (RCD) is a regulable cell death that involves well-organized signaling cascades and molecular mechanisms. RCD is implicated in fundamental processes such as organ production and tissue remodeling, removing superfluous structures or cells, and regulating cell numbers. Previous studies have not been able to reveal the complete mechanisms, and novel methods of RCD are constantly being proposed. Two metal ions, iron (Fe) and copper (Cu) are essential factors leading to RCDs that not only induce ferroptosis and cuproptosis, respectively but also lead to cell impairment and eventually diverse cell death. This review summarizes the direct and indirect mechanisms by which Fe and Cu impede cell growth and the various forms of RCD mediated by these two metals. Moreover, we aimed to delineate the interrelationships between these RCDs with the distinct pathways of ferroptosis and cuproptosis, shedding light on the complex and intricate mechanisms that govern cellular survival and death. Finally, the prospects outlined in this review suggest a novel approach for investigating cell death, which may involve integrating current therapeutic strategies and offer a promising solution to overcome drug resistance in certain diseases. Video Abstract.
Collapse
Affiliation(s)
- Yu Li
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, P.R. China
| | - Yuhui Du
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, P.R. China
| | - Yujie Zhou
- Basic Science Institute, Sungkyunkwan University, Suwon, South Korea
| | - Qianhui Chen
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Zhijie Luo
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yufan Ren
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xudan Chen
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Guoan Chen
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, P.R. China.
| |
Collapse
|
9
|
Teng Y, Gao L, Mäkitie AA, Florek E, Czarnywojtek A, Saba NF, Ferlito A. Iron, Ferroptosis, and Head and Neck Cancer. Int J Mol Sci 2023; 24:15127. [PMID: 37894808 PMCID: PMC10606477 DOI: 10.3390/ijms242015127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Ferroptosis is an iron-dependent regulatory form of cell death characterized by the accumulation of intracellular reactive oxygen species and lipid peroxidation. It plays a critical role not only in promoting drug resistance in tumors, but also in shaping therapeutic approaches for various malignancies. This review aims to elucidate the relationship between ferroptosis and head and neck cancer treatment by discussing its conceptual framework, mechanism of action, functional aspects, and implications for tumor therapy. In addition, this review consolidates strategies aimed at improving the efficacy of head and neck cancer treatment through modulation of ferroptosis, herein serving as a valuable reference for advancing the treatment landscape for this patient population.
Collapse
Affiliation(s)
- Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Lixia Gao
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China;
| | - Antti A. Mäkitie
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, FI-00014 Helsinki, Finland;
| | - Ewa Florek
- Laboratory of Environmental Research, Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland;
| | - Agata Czarnywojtek
- Department of Pharmacology, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland
| | - Nabil F. Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Alfio Ferlito
- Coordinator of the International Head and Neck Scientific Group, 35125 Padua, Italy;
| |
Collapse
|