1
|
Zeng C, Huang D, Wang L, Liang H, Ma X. Silencing ZIC5 suppresses glycolysis and promotes disulfidptosis in lung adenocarcinoma cells. Cancer Biol Ther 2025; 26:2501780. [PMID: 40366858 PMCID: PMC12080275 DOI: 10.1080/15384047.2025.2501780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 04/22/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025] Open
Abstract
OBJECTIVE This study aims to explore the effects of silencing Zic family member 5 (ZIC5) on glucose metabolism and disulfidptosis in lung adenocarcinoma (LUAD) cells. METHODS Data from The Cancer Genome Atlas (TCGA) was used to analyze ZIC5 expression in LUAD and its association with patient outcomes. ZIC5 was silenced in A549 and H1299 cells using siRNA. The expression of ZIC5 mRNA and protein was assessed by qRT-PCR and Western blot. Cell proliferation was evaluated through CCK-8 and 5-ethynyl-2'-deoxyuridine (EdU) assays, while glucose uptake, lactate production, and ATP levels were measured to assess energy metabolism. Seahorse XF analysis was used to evaluate extracellular acidification rate (ECAR) and oxygen consumption rate (OCR). Disulfidptosis was assessed through NADP+/NADPH ratio, glutathione (GSH) content, GSSG/GSH ratio, and immunofluorescence staining. RESULTS ZIC5 is highly expressed in LUAD and is associated with poor patient prognosis. Silencing ZIC5 significantly reduced its mRNA and protein levels in A549 and H1299 cells, markedly inhibited cell proliferation, and led to significant decreases in glucose uptake, lactate production, ATP levels, ECAR, and OCR. Additionally, silencing ZIC5 resulted in an increased NADP+/NADPH ratio, decreased GSH levels, and a reduced GSSG/GSH ratio, alongside classic disulfidptosis features. CONCLUSION ZIC5 plays a crucial role in promoting LUAD cell proliferation and energy metabolism while inhibiting disulfidptosis. Silencing ZIC5 markedly suppresses these processes, indicating its potential as a therapeutic target in LUAD.
Collapse
Affiliation(s)
- Cimei Zeng
- Department of Respiratory and Critical Care Medicine, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| | - Denggao Huang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| | - Lei Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| | - Haimei Liang
- Department of Respiratory and Critical Care Medicine, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| | - Ximiao Ma
- Thoracic Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| |
Collapse
|
2
|
Wang K, Li L, Liang G, Xiao H, Zhang L, Liu T. Sonodynamic activated nanoparticles with Glut1 inhibitor and cystine-containing polymer stimulate disulfidptosis for improved immunotherapy in bladder cancer. Biomaterials 2025; 319:123178. [PMID: 39978048 DOI: 10.1016/j.biomaterials.2025.123178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Disulfidptosis, a novel form of programmed cell death characterized by cystine accumulation and disulfide stress, primarily affects metabolically active tumors like bladder cancer, which is often considered to be a highly metabolic and energy-consuming tumor. However, translating disulfidptosis induction into clinical practice face substantial obstacles, including the limited solubility of key inducers, insufficient cystine buildup within cells, and cellular mechanisms regulating the NADP+/NADPH equilibrium. To fully unlock the therapeutic potential of disulfidptosis, a promising solution has emerged in the form of nanotechnology combined with sonodynamic therapy (SDT). This study reports a novel approach that enhances disulfidptosis through SDT, simultaneously promoting immunogenic cell death (ICD) and improving the immunosuppressive tumor microenvironment. The system, SPCP/CCP@Bay, comprises a degradable sonodynamic-pseudo-conjugate-polymer (SPCP) and a cystine-containing polymer (CCP), loaded with Bay-876. Following intravenous administration, SPCP/CCP@Bay effectively accumulates at tumor sites. Under ultrasound radiation, SPCP/CCP@Bay effectively releases Bay-876, disrupts the intracellular redox balance, releases cystine from CCP, and induces disulfidptosis. Moreover, SPCP/CCP@Bay induces ICD and synergizes with PD-1 monoclonal antibodies (α-PD-1) to suppress tumor growth. This integrated strategy holds significant promise in reshaping the tumor microenvironment, converting "cold tumors" to "hot tumors", and advancing the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Ke Wang
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Lin Li
- Department of Rehabilitation, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ganghao Liang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Lingpu Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Tao Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
3
|
Chen Y, Lin X, Qiu J, Sun Y, Wu B, Shang H, Deng L, Wang X, Li N, Huang C, Zhang T, Wu Z, Hou G, Yan X, Wang S, Cheng W. Ultrasound-responsive nanobubble-mediated sonodynamic therapy sensitizes disulfidptosis in the treatment of liver hepatocellular carcinoma. ULTRASONICS SONOCHEMISTRY 2025; 118:107368. [PMID: 40294549 PMCID: PMC12056780 DOI: 10.1016/j.ultsonch.2025.107368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/13/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025]
Abstract
Disulfidptosis, a newly identified regulated cell death, is linked to tumor progression, particularly in cancers with elevated SLC7A11 expression. This study investigates SLC7A11 expression in liver hepatocellular carcinoma (LIHC) and evaluates the therapeutic potential of ICG@C3F8-KL nanobubbles (NBs) combined with sonodynamic therapy (SDT) for inducing disulfidptosis. Bioinformatics analysis of TCGA datasets revealed upregulation of SLC7A11 in LIHC tissues. The synthesized ICG@C3F8-KL NBs exhibited a mean diameter of 156.46 nm and stable properties, with high encapsulation efficiencies of 51.32 % ± 0.7 % for KL and 80.15 % ± 0.21 % for ICG. In vitro, ICG@C3F8-KL NBs, under ultrasound, generated reactive oxygen species (ROS), enhancing cytotoxicity in HepG2 cells with an IC50 lower than KL alone. These NBs also inhibited cell migration and colony formation, suggesting disulfidptosis induction via altered glucose uptake and NADP+/NADPH ratio, as well as F-actin contraction. In vivo, ICG@C3F8-KL NBs accumulated in tumor tissues and suppressed growth without significant toxicity. Unsupervised clustering of disulfidptosis-related genes in TCGA LIHC cohort identified subtypes with distinct prognoses, and a predictive model based on five key genes was developed. In conclusion, ICG@C3F8-KL NBs, combined with ultrasound, effectively induce disulfidptosis, offering a promising strategy for LIHC treatment, with the potential for personalized therapy informed by disulfide-associated gene signatures.
Collapse
Affiliation(s)
- Yichi Chen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Xin Lin
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Jiayue Qiu
- Dr. Nesher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China
| | - Yucao Sun
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Bolin Wu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Haitao Shang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Liwen Deng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Xi Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Nanxing Li
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Chen Huang
- Dr. Nesher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China.
| | - Tianhong Zhang
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Zhiguang Wu
- School of Medicine and Healthcare, State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin 150080 China.
| | - Gang Hou
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Xiaohui Yan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361005, China
| | - Shoufeng Wang
- Qijing Machinery Co.,Ltd, Ningbo, Zhejiang 315600, China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China.
| |
Collapse
|
4
|
Wan J, Shi JH, Shi M, Huang H, Zhang Z, Li W, Guo C, Bao R, Yu X, Han Q, Du X, Li S, Ye Y, Cui X, Li X, Li JH, Zou Q. Lactate dehydrogenase B facilitates disulfidptosis and exhaustion of tumour-infiltrating CD8 + T cells. Nat Cell Biol 2025; 27:972-982. [PMID: 40461882 DOI: 10.1038/s41556-025-01673-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 04/16/2025] [Indexed: 06/19/2025]
Abstract
The aberrant accumulation of intracellular disulfides promotes cancer cell disulfidptosis; however, how disulfide stress influences tumour-infiltrating CD8+ T cell function remains unknown. Here we demonstrate that lactate dehydrogenase B (LDHB) facilitates intratumoural CD8+ T cell disulfidptosis and exhaustion, leading to impaired antitumour immunity. SLC7A11-mediated cystine uptake by CD8+ T cells induces disulfidptosis, which plays critical roles in the development of exhausted CD8+ T cells. LDHB restricts glucose-6-phosphate dehydrogenase (G6PD) activity in exhausted CD8+ T cells by interacting with G6PD, causing NADPH depletion and consequently triggering disulfidptosis. Accordingly, the loss of LDHB in T cells prevents disulfidptosis-dependent CD8+ T cell exhaustion and improves antitumour immunity. Mechanistically, STAT3 directs LDHB expression to limit G6PD activity and mediate disulfidptosis in exhausted CD8+ T cells. Our results highlight the distinct roles of disulfidptosis and ferroptosis in driving CD8+ T cell exhaustion and suggest a potential therapeutic strategy to target LDHB in cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Wan
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Hong Shi
- Central Laboratory, Hebei International Joint Research Center for Digital Twin Diagnosis and Treatment of Digestive Tract Tumors, Affiliated Hospital of Hebei University, Baoding, China
| | - Min Shi
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenyan Li
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenyue Guo
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rujuan Bao
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Yu
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiaoqiao Han
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xian Du
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Song Li
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youqiong Ye
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xia Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Jing-Hua Li
- Department of Hepatobiliary Surgery, Baoding Key Laboratory of Precision Diagnosis and Treatment of Digestive Tract Tumors, Affiliated Hospital of Hebei University, Baoding, China.
| | - Qiang Zou
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Li Q, Gan B. Disulfidptosis meets antitumour immunity. Nat Cell Biol 2025; 27:886-887. [PMID: 40473855 DOI: 10.1038/s41556-025-01679-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2025]
Affiliation(s)
- Qidong Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
6
|
Li S, He J, Kuang H, Wang X, Zhou M, Li D, Kang B, He H, He L, Lin W, Lv Y. Rab11a-dependent recycling of Glut3 inhibits seizure-induced neuronal disulfidptosis by alleviating glucose deficiency. Cell Biosci 2025; 15:69. [PMID: 40437641 PMCID: PMC12121293 DOI: 10.1186/s13578-025-01396-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/12/2025] [Indexed: 06/01/2025] Open
Abstract
Seizures can trigger neuronal glucose deficiency, thereby inducing disulfidptosis. Disulfidptosis is a novel cell death mechanism characterized by the abnormal accumulation of disulfide caused by glucose deficiency. However, the mechanism underlying disulfidptosis caused by glucose deficiency in seizures remains elusive. Rab11a-dependent recycling of glucose transporter 3 (Glut3) is closely related to glucose metabolism in neurons, which may contribute to neuronal disulfidptosis after seizures by abnormal glucose metabolism. So here we introduced a well-established in vitro model of seizures to evaluate cell survival, glucose levels, disulfidptosis biomarkers, Glut3 and Rab11a expression, the recycling ratio of Glut3, and the protein complex of Glut3-Rab11a. Cell survival rates and glucose levels were lower in the in vitro model of seizures, accompanied by elevated levels of disulfidptosis markers. Moreover, the surface expression and the recycling ratio of Glut3, as well as the protein complex of Glut3-Rab11a, were positively correlated with Rab11a expression. Lastly, Rab11 overexpression improved cell survival rates, increased glucose levels, and decreased the levels of disulfidptosis biomarkers in the in vitro model of seizure. Rab11a-dependent recycling of Glut3 inhibited seizure-induced neuronal disulfidptosis by alleviating glucose deficiency.
Collapse
Affiliation(s)
- Sijun Li
- Department of Geriatric rehabilitation/Clinical Research Center for Geriatric Disorders of Guangxi Zhuang Autonomous Region, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Junrui He
- Department of Geriatric rehabilitation/Clinical Research Center for Geriatric Disorders of Guangxi Zhuang Autonomous Region, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Huimin Kuang
- Department of Neurology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Xiaojuan Wang
- Department of Geriatric rehabilitation/Clinical Research Center for Geriatric Disorders of Guangxi Zhuang Autonomous Region, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Muhua Zhou
- Department of Geriatric rehabilitation/Clinical Research Center for Geriatric Disorders of Guangxi Zhuang Autonomous Region, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Dongmei Li
- Department of Geriatric rehabilitation/Clinical Research Center for Geriatric Disorders of Guangxi Zhuang Autonomous Region, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Baoren Kang
- Department of Geriatric rehabilitation/Clinical Research Center for Geriatric Disorders of Guangxi Zhuang Autonomous Region, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Honghu He
- Department of Geriatric rehabilitation/Clinical Research Center for Geriatric Disorders of Guangxi Zhuang Autonomous Region, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Lina He
- Department of Geriatric rehabilitation/Clinical Research Center for Geriatric Disorders of Guangxi Zhuang Autonomous Region, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Wei Lin
- Department of Geriatric rehabilitation/Clinical Research Center for Geriatric Disorders of Guangxi Zhuang Autonomous Region, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| | - Yuan Lv
- Department of Neurology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, No 85 Hedi Road, Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| |
Collapse
|
7
|
Yu W, Jin D, Zhang Y, Wang S, Yu J, Liu M, Dai Y, Yin Y, Cheng J, Liu Y. Provoking tumor disulfidptosis by single-atom nanozyme via regulating cellular energy supply and reducing power. Nat Commun 2025; 16:4877. [PMID: 40419525 DOI: 10.1038/s41467-025-60015-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
Disulfidptosis, a recently identified form of programmed cell death, is initiated by depletion of endogenous nicotinamide adenine dinucleotide phosphate (NADPH) under glucose starvation. Tumor cells, owing to their heightened requirements of energy and nutrients, are more susceptible to disulfidptosis than normal cells. Here, we introduced an effective strategy to induce tumor disulfidptosis via interrupting cellular energy supply and reducing power by integrating a copper single-atom nanozyme (CuSAE) and glucose oxidase (GOx). GOx induces glucose starvation, impeding generation of NADPH through pentose phosphate pathway (PPP). CuSAE mimics NADPH oxidase, depleting existing NADPH, which intensifies the blockade of disulfide reduction and efficiently triggers disulfidptosis of tumor cells. Furthermore, CuSAE exhibits peroxidase- and glutathione oxidase-mimicking activities, catalyzing generation of •OH radical and depletion cellular GSH, which enhances oxidative stress and exacerbates cell damage. Disulfidptosis is confirmed as the predominant type of cell death induced by GOx/CuSAE. In vivo assays demonstrated the high antitumor potency of GOx/CuSAE in treating with female tumor-bearing mice, with minimal systemic toxicity observed. This work introduces a promising strategy for designing antitumor agents by inducing disulfidptosis. The enzyme hybrids that combine nanozymes and natural enzymes offer a feasible approach to achieve this multifaceted therapeutic goal.
Collapse
Affiliation(s)
- Wenxin Yu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, China
| | - Duo Jin
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, China
| | - Yajie Zhang
- Central Laboratory, Department of Biobank, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Shenghu Wang
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, China
| | - Jiaji Yu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, China
| | - Manman Liu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, China
| | - Yi Dai
- College of Pharmaceutical Sciences, Anhui Xinhua University, Hefei, China
| | - Yichen Yin
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, China
| | - Junjie Cheng
- Department of Radiology, Zhongda Hospital, Medical School; Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China.
| | - Yangzhong Liu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China.
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
8
|
Zhu J, Zhu X, Su T, Zhou H, Wang S, Shi W. The Development and Assessment of a Unique Disulfidptosis-Associated lncRNA Profile for Immune Microenvironment Prediction and Personalized Therapy in Gastric Adenocarcinoma. Biomedicines 2025; 13:1224. [PMID: 40427051 PMCID: PMC12109475 DOI: 10.3390/biomedicines13051224] [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: 03/13/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) are crucial factors affecting the occurrence, progression, and prognosis of gastric carcinoma (GC). The accumulation of disulfide bonds to excessive levels in cells expressing high SLC7A11 triggers disulfidptosis, which functions as a regulated form of cellular death. Research has demonstrated that upregulated SLC7A11 is common in human cancers, but the effect of disulfidptosis on GC remains unclear. Identifying lncRNAs associated with disulfidptosis (drlncRNAs) and establishing a prognostic risk profile holds considerable importance for advancing GC research and treatment. Methods: Clinical records and transcriptomic datasets from individuals with GC were acquired from The Cancer Genome Atlas (TCGA) repository. A three-drlncRNA risk model was built using three common regression analysis methods. Then we used receiver operating characteristic (ROC) curves, independent prognostic analysis, and additional statistical approaches to assess the precision of the model. This investigation additionally encompassed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, immune cell infiltration evaluation, and pharmacological sensitivity predictions. To further investigate immunotherapy response disparities between patient cohorts with elevated- and reduced-risk scores, analyses of tumor mutational burden (TMB), tumor immune dysfunction and exclusion (TIDE), and microsatellite instability (MSI) were implemented. Results: We constructed a unique model composed of three drlncRNAs (AC107021.2, AC016394.2, and AC129507.1). Its independent prognostic capability for GC patients was validated through both single-variable and multivariable Cox regression analyses. GO and KEGG pathway assessments revealed predominant enrichment within the elevated-risk cohort, particularly in pathways involving sulfur compound interactions, traditional Wnt signaling mechanisms, cell-substrate adherens junctions, and cAMP signaling cascades, among others. Tumor microenvironment (TME) evaluation demonstrated elevated ImmuneScores, StromalScores, and ESTIMATEScores within the high-risk patient population. Concurrently, this elevated-risk cohort exhibited enhanced immune cell infiltration patterns, whereas the reduced-risk group displayed superior expression of immune checkpoints (ICPs). Additional investigations revealed that patients categorized into the reduced-risk classification possessed greater tumor mutational burden, increased MSI-high proportions, and diminished tumor immune dysfunction and exclusion scores compared to their high-risk counterparts. Pharmacological sensitivity assessments confirmed the superior efficacy of several therapeutic agents, including gemcitabine and veliparib (ABT.888), in patients with lower risk classifications. Conclusions: Our established risk stratification system demonstrates independent prognostic predictive capacity while offering personalized clinical intervention guidance for individuals diagnosed with GC.
Collapse
Affiliation(s)
- Jiyue Zhu
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| | - Xiang Zhu
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| | - Tingting Su
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
| | - Huiqing Zhou
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China;
| | - Shouhua Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| | - Weibin Shi
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| |
Collapse
|
9
|
Gou ZH, Su N, Li XC, Ren DP, Ren SS, Wang L, Wang Y. Integrated transcriptomic and immune profiling reveals crucial molecular pathways and hub genes associated with postoperative delirium in elderly patients. Front Med (Lausanne) 2025; 12:1580355. [PMID: 40417694 PMCID: PMC12098571 DOI: 10.3389/fmed.2025.1580355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/15/2025] [Indexed: 05/27/2025] Open
Abstract
Background Postoperative delirium (POD) manifests as severe mental disorientation, often experienced by elderly patients undergoing surgery, significantly hindering recovery and deteriorating the quality of life. Despite numerous clinical studies, the molecular mechanisms behind POD in elderly patients are still not well understood, requiring further investigation to identify potential biomarkers and therapeutic targets. Methods This study amalgamates Gene Set Variation Analysis (GSVA), Weighted Gene Co-expression Network Analysis (WGCNA), differential expression analysis, and immune infiltration assessments to identify molecular pathways and hub genes linked to the initiation of POD in the elderly. Gene expression data were sourced from the GSE163943 dataset in the Gene Expression Omnibus (GEO) database. A total of 18,894 protein-coding genes were extracted for analysis. Results We constructed a gene co-expression network using WGCNA and performed GSVA to investigate the link between POD and different types of cell death. The results indicated that POD is positively associated with pyroptosis and parthanatos, while negatively correlated with oxidative stress and disulfidptosis. Differential expression analysis revealed 145 differentially expressed genes (DEGs), including 83 downregulated and 62 upregulated genes. Analysis of functional enrichment revealed that DEGs were enriched in activities like neuron projection development, axonogenesis, and synapse organization, with KEGG pathway analysis identifying neuroactive ligand-receptor interaction and neurodegeneration pathways. Gene Set Enrichment Analysis (GSEA) further revealed the upregulation of the apoptosis pathway and the downregulation of neuroactive ligand-receptor interaction. Protein-protein interaction (PPI) network analysis identified 10 hub genes, including COL18A1, CD63, and LTF. Immune infiltration analysis indicated that the occurrence of POD is strongly associated with immune cell activation, particularly in T cells and macrophages. Conclusion Overall, this research primarily examines the intricate interplay between cell death processes and alterations in the immune microenvironment throughout the development of geriatric POD, pinpointing essential genes that provide vital theoretical support for further studies on geriatric POD. However, this discovery is only an initial one derived from analyzing the datasets. Upcoming research ought to evaluate and scrutinize additional datasets and conduct essential experiments to guarantee the precision and widespread relevance of the analytical findings.
Collapse
Affiliation(s)
- Zi-han Gou
- Department of Anesthesiology, The People’s Hospital of kaizhou District Chongqing, Chongqing, China
| | - Nan Su
- Inner Mongolia People’s Hospital Department of Surgical Anesthesia, Inner Mongolia, China
| | - Xiao-chuan Li
- Department of Orthopaedic, Chongqing Sanbo Changan Hospital, Chongqing, China
| | - Da-peng Ren
- Department of Anesthesiology, The People’s Hospital of kaizhou District Chongqing, Chongqing, China
| | - Shan-shan Ren
- Department of Anesthesiology, The Thirteenth People’s Hospital of Chongqing, Chongqing, China
| | - Lin Wang
- Department of Anesthesiology, The Thirteenth People’s Hospital of Chongqing, Chongqing, China
| | - Yao Wang
- Department of Anesthesiology, The Thirteenth People’s Hospital of Chongqing, Chongqing, China
| |
Collapse
|
10
|
Kumari R, Banerjee S. Regulation of Different Types of Cell Death by Noncoding RNAs: Molecular Insights and Therapeutic Implications. ACS Pharmacol Transl Sci 2025; 8:1205-1226. [PMID: 40370994 PMCID: PMC12070317 DOI: 10.1021/acsptsci.4c00681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
Noncoding RNAs (ncRNAs) are crucial regulatory molecules in various biological processes, despite not coding for proteins. ncRNAs are further divided into long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) based on the size of their nucleotides. These ncRNAs play crucial roles in transcriptional, post-transcriptional, and epigenetic regulation. The regulatory roles of noncoding RNAs, including lncRNAs, miRNAs, and circRNAs, are essential in various modalities of cellular death, such as apoptosis, ferroptosis, cuproptosis, pyroptosis, disulfidptosis, and necroptosis. These noncoding RNAs are integral to modulating gene expression and protein functionality during cellular death mechanisms. In apoptosis, lncRNAs, miRNAs, and circRNAs influence the transcription of apoptotic genes. In ferroptosis, these noncoding RNAs target genes and proteins involved in iron homeostasis and oxidative stress responses. For cuproptosis, noncoding RNAs regulate pathways associated with the accumulation of copper ions, leading to cellular death. During pyroptosis, noncoding RNAs modulate inflammatory mediators and caspases, affecting the proinflammatory cell death pathway. In necroptosis, noncoding RNAs oversee the formation and functionality of necrosomes, thereby influencing the balance between cellular survival and death. Disulfidptosis is a unique type of regulated cell death caused by the excessive formation of disulfide bonds within cells, leading to cytoskeletal collapse and oxidative stress, especially under glucose-limited conditions. This investigation highlights the complex mechanisms through which noncoding RNAs coordinate cellular death, emphasizing their therapeutic promise as potential targets, particularly in the domain of cancer treatment.
Collapse
Affiliation(s)
- Reshmi Kumari
- Department of Biotechnology, School
of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Satarupa Banerjee
- Department of Biotechnology, School
of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
11
|
Shang N, Wang J, Liu Z, Wang Y, Zhang D, Liu H, Zhang Y, Dai G, Guan X. Integrated Analysis of Disulfidptosis-Related Genes Identifies CD2AP as a Potential Therapeutic Target for Hepatocellular Carcinoma. Int J Mol Sci 2025; 26:4454. [PMID: 40362690 PMCID: PMC12072785 DOI: 10.3390/ijms26094454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly cancer with limited treatment options for patients at advanced stages. It is urgent to develop reliable prognostic risk models and identify more biomarkers to improve the clinical outcomes of patients with HCC. Disulfidptosis is a newly discovered form of regulated cell death (RCD), and research on the comprehensive roles of disulfidptosis-related genes (DRGs) in HCC prognosis and development remains limited. In this paper, we systematically analyzed the expression levels and prognostic profiles of 26 DRGs in HCC samples from The Cancer Genome Atlas (TCGA) cohort and developed a prognostic risk model using seven hub DRGs. The independent prognostic value of the risk model was further validated in the external cohort. The overall survival of patients with HCC in the low-risk group was significantly longer than that of those in the high-risk group. Subsequently, the protein level of CD2-associated protein (CD2AP) was found to be highly expressed in HCC clinical tissues and associated with the severity of HCC. In vitro experiments demonstrated that the down-regulation of CD2AP attenuated the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) abilities of HCC cells. Taken together, our study revealed that the DRG CD2AP may serve as a potential biomarker for HCC and offer support for prognosis prediction of patients with HCC.
Collapse
Affiliation(s)
- Ning Shang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Jianwei Wang
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450001, China;
| | - Zihan Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Yake Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Di Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Huanfei Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Yaqing Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Guifu Dai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Xiaowen Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| |
Collapse
|
12
|
Yang W, Ma K, Yin S, Wang W, An H, Huang Y, Guo H, Ao L, Yang Z, Zhang F. Multiomic Landscape of Primary Hypothyroidism Induced by Subchronic Exposure to Low-Dose Novel PFOS Substitute OBS in Human and Murine Models. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:8329-8344. [PMID: 40178189 DOI: 10.1021/acs.est.4c10565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Sodium p-perfluorous nonenoxybenzenesulfonate (OBS) as a novel surrogate for perfluorooctanesulfonate (PFOS) has been extensively utilized in industrial manufacturing and daily life. However, studies on OBS-induced environmental health risks of obstructive biosynthesis (OBS) are currently limited, particularly the risk for thyroid diseases. Following the construction of in vivo (mouse) and in vitro (normal human primary thyrocytes) models of subchronic low-dose OBS exposure, we explored the thyroid-disrupting effects of OBS through multiomics approaches and experimental validations. Our results showed that subchronic exposure to low doses of OBS led to primary hypothyroidism in mice, presenting with reduced number and functional abnormalities of thyrocytes. Further in vitro assays confirmed that low-dose OBS-induced disulfidptosis, a newly discovered form of programmed cell death, in human primary thyrocytes. Meanwhile, exposure to low-dose OBS remarkably suppressed thyroid hormone synthesis pathways in mouse and human thyrocytes. The charted multiomic landscape of OBS-induced primary hypothyroidism in mammals revealed the thyroid toxicity and endocrine-disrupting properties of OBS, suggesting that it is not a safe alternative to PFOS.
Collapse
Affiliation(s)
- Wang Yang
- Chongqing Specialized Center for Diagnosis and Treatment of Thyroid Nodules and Thyroid Cancer, Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ke Ma
- Chongqing Specialized Center for Diagnosis and Treatment of Thyroid Nodules and Thyroid Cancer, Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
- Clinical Medical College, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Supeng Yin
- Chongqing Specialized Center for Diagnosis and Treatment of Thyroid Nodules and Thyroid Cancer, Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
| | - Weihua Wang
- Chongqing Specialized Center for Diagnosis and Treatment of Thyroid Nodules and Thyroid Cancer, Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
| | - Huihui An
- Medical Research Center, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
| | - Yinde Huang
- Chongqing Specialized Center for Diagnosis and Treatment of Thyroid Nodules and Thyroid Cancer, Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
| | - Hong Guo
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
| | - Lin Ao
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zeyu Yang
- Chongqing Specialized Center for Diagnosis and Treatment of Thyroid Nodules and Thyroid Cancer, Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
| | - Fan Zhang
- Chongqing Specialized Center for Diagnosis and Treatment of Thyroid Nodules and Thyroid Cancer, Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China
- Clinical Medical College, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| |
Collapse
|
13
|
Wan S, Liang C, Wu C, Wang S, Wang J, Xu L, Zhang X, Hou Y, Xia Y, Xu L, Huang X. Disulfidptosis in tumor progression. Cell Death Discov 2025; 11:205. [PMID: 40295497 PMCID: PMC12038022 DOI: 10.1038/s41420-025-02495-9] [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: 11/27/2024] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
Disulfidptosis, a regulated cell death modality driven by the cystine transporter solute carrier family 7 member 11 (SLC7A11), is characterized by actin cytoskeleton collapse under glucose starvation. This review systematically elucidates the pivotal role of disulfidptosis in tumor metabolic reprogramming, with a focus on its molecular mechanisms and distinctions from other cell death pathways. The core mechanisms include SLC7A11-mediated cystine overload and NRF2/c-Myc-regulated pentose phosphate pathway activation. By integrating multiomics data and single-cell transcriptomics, we comprehensively decipher the heterogeneous expression patterns of disulfidptosis-related genes (DRGs) and their dynamic interplay with immune microenvironment remodeling. Furthermore, the coexpression networks of DRGs and disulfidptosis-related long noncoding RNAs (DRLs) offer novel insights into tumor diagnosis, prognosis, and targeted therapy. Therapeutically, SLC7A11 inhibitors (e.g., HG106) and glucose transporter inhibitors (e.g., BAY-876) demonstrate efficacy by exploiting metabolic vulnerabilities, whereas natural compounds synergizing with immune checkpoint blockade provide strategies to counteract immunosuppressive microenvironments. Through interdisciplinary collaboration and clinical translation, disulfidptosis research holds transformative potential in redefining precision oncology.
Collapse
Affiliation(s)
- Senlin Wan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Changming Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Chengwei Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Song Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Jiawei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Lishuai Xu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Xu Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Yinfen Hou
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Yabin Xia
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Li Xu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Xiaoxu Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China.
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China.
| |
Collapse
|
14
|
Zhen W, Zhao T, Chen X, Zhang J. Unlocking the Potential of Disulfidptosis: Nanotechnology-Driven Strategies for Advanced Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500880. [PMID: 40269657 DOI: 10.1002/smll.202500880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Tumor tissues exhibit elevated oxidative stress, with the cystine-glutamate transporter xCT solute carrier family 7 member 11 (xCT/SLC7A11) protecting cancer cells from oxidative damage by facilitating cystine uptake for glutathione synthesis. Disulfidptosis, a newly identified form of programmed cell death (PCD), occurs in cells with high xCT/SLC7A11 expression under glucose-deprived conditions. Distinct from other PCD pathways, disulfidptosis is characterized by aberrant disulfide bond formation and cellular dysfunction, ultimately resulting in cancer cell death. This novel mechanism offers remarkable therapeutic potential by targeting the inherent oxidative stress vulnerabilities of rapidly growing cancer cells. Advances in nanotechnology enable the development of nanomaterials capable of inducing reactive oxygen species (ROS) generation, disrupting disulfide bonds. In addition, they are capable to deliver therapeutic agents directly to tumors, thereby improving therapeutic precision and minimizing off-target effects. Moreover, combining disulfidptosis with ROS-induced immunogenic cell death can remodel the tumor microenvironment and enhance anti-tumor immunity. This review explores the mechanisms underlying disulfidptosis, its therapeutic potential in cancer treatment, and the synergistic role of nanotechnology in amplifying its effects. Selective induction of disulfidptosis using nanomaterials represents a promising strategy for achieving more effective, selective, and less toxic cancer therapies.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Centre of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 138667, Singapore
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Centre of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 138667, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117575, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117575, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Centre of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Centre of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 138667, Singapore
| |
Collapse
|
15
|
Li J, Jiang Y, Nong S, Liang L, Chen L, Gong Q. Development of a machine learning-derived programmed cell death index for prognostic prediction and immune insights in colorectal cancer. Discov Oncol 2025; 16:608. [PMID: 40274671 PMCID: PMC12021754 DOI: 10.1007/s12672-025-02323-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Colorectal cancer (CRC) is a major contributor to cancer-related mortality worldwide, emphasizing the need for improved prognostic tools and therapeutic strategies. Programmed cell death, encompassing diverse modalities, plays a critical role in tumor biology and therapy response. Utilizing machine learning techniques, we developed a novel Programmed Cell Death Index (PCDI) incorporating multiple forms of PCD-related genes to predict outcomes in colorectal cancer CRC patients. The PCDI demonstrated robust prognostic performance, stratifying patients into high- and low-risk groups across multiple cohorts, with high PCDI scores correlating with poor survival, advanced tumor stage, and aggressive pathological features. A nomogram integrating PCDI with clinical variables showed strong predictive accuracy for 1-, 3-, and 5 year survival rates. Functional analysis revealed significant metabolic differences between high- and low-PCDI groups. Immune profiling identified associations between PCDI and immunosuppressive microenvironments, including elevated regulatory T cell levels and reduced PD-L1 expression in high-PCDI patients. Patients with high PCDI exhibited a potential resistance to immune checkpoint inhibitors. These findings emphasize PCDI's potential as a prognostic biomarker and a tool for guiding personalized therapeutic strategies in CRC patients.
Collapse
Affiliation(s)
- Jinping Li
- Yangzhou Polytechnic College, Yangzhou, 225009, Jiangsu, China
| | - Yan Jiang
- Department of Nephrology, Youjiang Medical College for Nationalities Affiliated Hospital, Baise, 533000, China
- Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi (Cultivation), Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Shengbin Nong
- Department of Nephrology, Youjiang Medical College for Nationalities Affiliated Hospital, Baise, 533000, China
- Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi (Cultivation), Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Liudan Liang
- Department of Nephrology, Youjiang Medical College for Nationalities Affiliated Hospital, Baise, 533000, China
- Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi (Cultivation), Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Liangchao Chen
- Department of Oncology, Xichong People's Hospital, Nanchong, 637200, China.
| | - Qiming Gong
- Department of Nephrology, Youjiang Medical College for Nationalities Affiliated Hospital, Baise, 533000, China.
- Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi (Cultivation), Youjiang Medical University for Nationalities, Baise, 533000, China.
| |
Collapse
|
16
|
Gujar V, Li H, Paull TT, Neumann CA, Weyemi U. Unraveling the nexus: Genomic instability and metabolism in cancer. Cell Rep 2025; 44:115540. [PMID: 40208791 PMCID: PMC12043202 DOI: 10.1016/j.celrep.2025.115540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025] Open
Abstract
The DNA-damage response (DDR) is a signaling network that enables cells to detect and repair genomic damage. Over the past three decades, inhibiting DDR has proven to be an effective cancer therapeutic strategy. Although cancer drugs targeting DDR have received approval for treating various cancers, tumor cells often develop resistance to these therapies, owing to their ability to undergo energetic metabolic reprogramming. Metabolic intermediates also influence tumor cells' ability to sense oxidative stress, leading to impaired redox metabolism, thus creating redox vulnerabilities. In this review, we summarize recent advances in understanding the crosstalk between DDR and metabolism. We discuss combination therapies that target DDR, metabolism, and redox vulnerabilities in cancer. We also outline potential obstacles in targeting metabolism and propose strategies to overcome these challenges.
Collapse
Affiliation(s)
- Vaibhavi Gujar
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haojian Li
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tanya T Paull
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Carola A Neumann
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Urbain Weyemi
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
17
|
Wang D, Wang C, Liu H, Zhang Z, Li M, Ge X, Bi A, Gao C, Tian X, Liu K, Zhu Z. Integrated bioinformatic analysis of immune infiltration and disulfidptosis related gene subgroups in type A aortic dissection. Sci Rep 2025; 15:13719. [PMID: 40258895 PMCID: PMC12012055 DOI: 10.1038/s41598-025-98149-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 04/09/2025] [Indexed: 04/23/2025] Open
Abstract
Type A aortic dissection (TAAD) is a lethal cardiovascular disease characterized by the separation of the layers within the aortic wall. The underlying pathological mechanisms of TAAD requires further elucidation to develop effective prevention and pharmacological treatment strategies. Inflammation plays a crucial role in TAAD pathogenesis. Disulfidptosis, an emerging type of cell death, may shed light on disease mechanisms. This study investigates the role of disulfidptosis-related genes in immune infiltration in TAAD. TAAD gene expression datasets were obtained from the Gene Expression Omnibus (GEO) database. Immune cell infiltration analysis assessed immune cell dysregulation in TAAD. Differentially expressed genes (DEGs) between TAAD samples and controls were identified and intersected with known disulfidptosis-related gene sets to obtain relevant DEGs. Hub genes were identified using machine learning algorithms. A diagnostic model was constructed using Least Absolute Shrinkage and Selection Operator (LASSO) regression on 25 TAAD samples. Consensus clustering classified TAAD samples based on disulfidptosis-related gene expression. Functional enrichment analyses, including Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, elucidated associated biological processes and pathways. A total of 13,316 DEGs were identified, among which 11 disulfidptosis-related genes were screened: INF2, CD2AP, PDLIM1, ACTN4, MYH10, MYH9, FLNA, FLNB, TLN1, MYL6, ACTB, CAPZB, DSTN, and IQGAP1. Most of these genes exhibited lower expression levels in TAAD samples, except CAPZB, and were correlated with immune cell infiltration. Cluster-specific DEGs were found in one cluster, involving several immune response processes. Co-clustering analysis based on disulfidptosis-related genes classified TAAD samples into two clusters, with higher gene expression levels observed in cluster C2 compared to cluster C1. Three key hub genes were identified, and potential therapeutic mechanisms for TAAD were explored. Immuno-infiltration results revealed significant differences in immune profiles, with higher immunological scores and more extensive immune infiltration in TAAD. Disulfidptosis occurs in TAAD and is associated with immune cell infiltration and metabolic activity, influencing immune cell function and responses. These findings suggest that disulfidptosis may promote TAAD progression through the induction of immune responses and metabolic activities. This research provides new insights into the pathogenesis and identifies potential therapeutic targets for TAAD.
Collapse
Affiliation(s)
- Duo Wang
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Chengwen Wang
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Haoyue Liu
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Zhenzhu Zhang
- Graduate School, Chengde Medical University, Baoding, China
| | - Mixia Li
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Xinpeng Ge
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Anqi Bi
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | | | - Xue Tian
- Jining Public Health Medical Center, Jining, China
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China.
| | - Zhicheng Zhu
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China.
| |
Collapse
|
18
|
Zhang M, Zheng H, Zhu X, Liu S, Jin H, Chen Y, Wan L, Zhang S, Zhang H. Synchronously Evoking Disulfidptosis and Ferroptosis via Systematical Glucose Deprivation Targeting SLC7A11/GSH/GPX4 Antioxidant Axis. ACS NANO 2025; 19:14233-14248. [PMID: 40178511 DOI: 10.1021/acsnano.5c00730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Disulfidptosis and ferroptosis are recently identified programmed cell deaths for tumor therapy, both of which highly depend on the intracellular cystine/cysteine transformation on the cystine transporter solute carrier family 7 member 11/glutathione/glutathione peroxidase 4 (SLC7A11/GSH/GPX4) antioxidant axis. However, disulfidptosis and ferroptosis are usually asynchronous due to the opposite effect of cystine transport on them. Herein, systematic glucose deprivation, by both inhibiting upstream glucose uptake and promoting downstream glucose consumption, is proposed to synchronously evoke disulfidptosis and ferroptosis. As an example, Au nanodots and Fe-apigenin (Ap) complexes coloaded FeOOH nanoshuttles (FeOOH@Fe-Ap@Au NSs) are employed to regulate the SLC7A11/GSH/GPX4 axis for performing disulfidptosis- and ferroptosis-mediated tumor therapy synchronously. In this scenario, Au nanodots exhibit glucose oxidase-like activity when consuming massive glucose. Meanwhile, Ap can inhibit glucose uptake by downregulating glucose transporter 1, depriving glucose fundamentally. The systematical glucose deprivation limits the supplement of NADPH and suppresses cystine/cysteine transformation on the SLC7A11/GSH/GPX4 axis, thus solving the contradiction of cystine transport on disulfidptosis and ferroptosis. In addition, the efficient delivery of exogenous iron ions by FeOOH@Fe-Ap@Au NSs and self-supplied H2O2 through Au nanodots-catalytic glucose oxidation facilitate intracellular Fenton reaction and therewith help to amplify ferroptosis. As a result of synchronous occurrence of disulfidptosis and ferroptosis, FeOOH@Fe-Ap@Au NSs exhibit good efficacy in an ovarian cancer therapeutic model.
Collapse
Affiliation(s)
- Mengsi Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
- Department of Gynecological Oncology, Gynecology and Obstetrics Center, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Hao Zheng
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Xuanqi Zhu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Shuwei Liu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Hao Jin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yang Chen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Lanlan Wan
- Department of Anesthesia, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Songling Zhang
- Department of Gynecological Oncology, Gynecology and Obstetrics Center, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Hao Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
19
|
Lin Z, Lv J, Dai C, Zhai Y, Jiang J, Gao Y, Li R, Fan J, Yu Y, Wu L, Yang Y. Identification of the disulfidptosis-related key gene CD2AP as a potential biomarker and new therapeutic target for LUAD patients by comprehensive multi-omics analysis. Discov Oncol 2025; 16:515. [PMID: 40214854 PMCID: PMC11992263 DOI: 10.1007/s12672-025-02308-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Lung Adenocarcinoma (LUAD) is a major subtype of Non-Small Cell Lung Cancer (NSCLC) with poor prognosis. Despite advances in molecular targeted therapy and immunotherapy, the five-year survival rate remains low. Disulfidptosis, a novel cell death mechanism, may play a role in tumor progression. CD2AP (CD2-associated protein), a key gene related to Disulfidptosis, is involved in cytoskeleton reorganization and signaling. This study aimed to explore CD2AP's function in LUAD and its potential as a biomarker and therapeutic target through multi-omics analysis. METHODS We analyzed CD2AP expression and clinical significance in LUAD using data from TCGA, GEO, and other public databases. We employed transcriptomics, methylation analysis, immune infiltration assays, and spatial transcriptomics. Kaplan-Meier survival analysis was used to assess the relationship between CD2AP expression and prognosis. Enrichment analysis identified biological processes and pathways related to CD2AP, while its association with the immune microenvironment and drug sensitivity was also evaluated. RESULTS CD2AP was significantly overexpressed in LUAD, and high expression correlated with poorer prognosis, including overall survival and progression-free survival. Enrichment analysis showed CD2AP is involved in cell adhesion, PI3K-Akt signaling, and immune escape, suggesting it promotes LUAD progression through these pathways. High CD2AP expression was associated with alterations in the tumor immune microenvironment and drug sensitivity, particularly to chemotherapeutics like Cisplatin, Etoposide, and Paclitaxel, and resistance to targeted therapies like Gefitinib. Spatial transcriptomics revealed higher CD2AP expression in tumor regions, especially in malignant cell-enriched areas. CONCLUSION This study highlights CD2AP's critical role in LUAD, particularly in immune microenvironment modulation, metabolic reprogramming, and drug response. CD2AP's high expression is linked to poor prognosis and may serve as a potential target for immunotherapy and drug response prediction.
Collapse
Affiliation(s)
- Zhiwu Lin
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Jing Lv
- Department of Orthopedics, Ziyang Central Hospital, Ziyang, 641300, China
| | - Chuanqiang Dai
- Department of Orthopedics, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yuanwei Zhai
- Department of Medical Imaging, Ziyang Central Hospital, Ziyang, 641300, China
| | - Jiudong Jiang
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yang Gao
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Rulin Li
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Jiangang Fan
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yang Yu
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Liang Wu
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yanchun Yang
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China.
| |
Collapse
|
20
|
Liu Y, Stockwell BR, Jiang X, Gu W. p53-regulated non-apoptotic cell death pathways and their relevance in cancer and other diseases. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00842-3. [PMID: 40204927 DOI: 10.1038/s41580-025-00842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Programmed cell death is a mechanism that is crucial for numerous physiological and pathological processes. Whereas p53-mediated apoptosis is a major cell death pathway in cancer, accumulating evidence indicates that p53 also has crucial roles in controlling different non-apoptotic cell death (NACD) pathways, including ferroptosis, necroptosis, pyroptosis, autophagy-dependent cell death, entotic cell death, parthanatos and paraptosis, and may regulate PANoptosis, cuproptosis and disulfidptosis. Notably, the function of p53 in these NACDs substantially contributes to its biological effects, particularly in cancer development and other pathological processes. In this Review, we discuss recent advances in understanding the roles and underlying mechanisms of p53-mediated NACDs, focusing on ferroptosis, necroptosis and pyroptosis. We discuss the complex and distinct physiological settings in which NACDs are regulated by p53, and potential targeting of p53-regulated NACDs for the treatment of cancer and other human diseases. Finally, we highlight several important questions concerning p53-regulated NACDs that warrant further investigation.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, USA
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
21
|
Li Q, Zhuang L, Gan B. Disulfidptosis. Curr Biol 2025; 35:R241-R243. [PMID: 40199244 DOI: 10.1016/j.cub.2025.02.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Li et al. introduce disulfidptosis, a recently identified form of regulated cell death triggered by excessive disulfide stress.
Collapse
Affiliation(s)
- Qidong Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Wu T, Liu K, Chen S, Ye Z, Xia J, He J, Xing P, Yang J, Qian Y, Chen M. Pulmonary microbiota disruption by respiratory exposure to carbon quantum dots induces neuronal damages in mice. JOURNAL OF HAZARDOUS MATERIALS 2025; 487:137255. [PMID: 39832471 DOI: 10.1016/j.jhazmat.2025.137255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/03/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Given the fact that carbon quantum dots (CQDs) have been commercially produced in quantities, it is inevitable to make their ways into environment and interact closely with the public. Even though CQDs in the environment have been reported to damage the central nervous system, the underlying mechanisms of neurotoxic effects of CQDs following respiratory exposure is still not clear. Intranasal instilled CQDs, mimicking respiratory exposure, induces neurobehavioral impairments associated with neuronal cell death of ferroptosis and disulfidptosis that is regulated by metabolic reprogramming of glutathione and cysteine pathways in the cortex and hippocampus where CQDs were hardly accumulated. Therefore, further exploration found that dysbiosis in the lung microbiome was found specifically manipulated by CQDs, which correlated with systemic and neuroinflammatory responses, implicating a lung-brain axis other than gut-brain axis as a critical pathway through which microbiota dysbiosis may impact neurological health after respiratory exposure to CQDs. This study pioneers the exploration of the neurological consequences of inhaled CQDs in the environment through the regulation of microbiome-lung-brain axis, which is key in understanding the mechanistic link between CQDs exposure and neurotoxicity. The findings could develop potential strategies for mitigating the neurological effects of CQDs even other types of nanoparticles.
Collapse
Affiliation(s)
- Tianshu Wu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China.
| | - Kehan Liu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China
| | - Siyuan Chen
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China
| | - Zongjian Ye
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China
| | - Jieyi Xia
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China
| | - Jing He
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China
| | - Pengcheng Xing
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China
| | - Jiafu Yang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China
| | - Yijing Qian
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China
| | - Min Chen
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, Nanjing 210009, China; School of Public Health, Southeast University, Nanjing 210009, China; Yancheng Kindergarten Teachers College, Yancheng 224005, China
| |
Collapse
|
23
|
Ye K, Han X, Tian M, Liu L, Gao X, Xia Q, Wang D. Analysis of human brain RNA-seq data reveals combined effects of 4 types of RNA modifications and 18 types of programmed cell death on Alzheimer's disease. J Transl Med 2025; 23:396. [PMID: 40181382 PMCID: PMC11969709 DOI: 10.1186/s12967-025-06324-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND RNA modification plays a critical role in Alzheimer's disease (AD) by modulating the expression and function of AD-related genes, thereby affecting AD occurrence and progression. Programmed cell death is closely related to neuronal death and associated with neuronal loss and cognitive function changes in AD. However, the mechanism of their joint action on AD remains unknown and requires further exploration. METHODS We used the MSBB RNA-seq dataset to analyze the correlation between RNA modification, programmed cell death, and AD. We used combined studies of RNA modification and programmed cell death to distinguish subgroups of patients, and the results highlight the strong correlation between RNA modification-related programmed cell death and AD. A weighted gene co-expression network was constructed, and the pivotal roles of programmed cell death genes in key modules were identified. Finally, by combining unsupervised consensus clustering, gene co-expression networks, and machine learning algorithms, an RNA modification-related programmed cell death network was constructed, and the pivotal roles of programmed cell death genes in key modules were identified. An RNA modification-related programmed cell death risk score was calculated to predict the occurrence of AD. RESULTS RPCD-related genes classified patients into subgroups with distinct clinical characteristics. Nineteen key genes were identified and an RPCD risk score was constructed based on the key genes. This score can be used for the diagnosis of AD and the assessment of disease progression in patients. The diagnostic efficacy of the RPCD risk score and the key genes was validated in the ROSMAP, GEO, and ADNI datasets. CONCLUSION This study uncovered that RNA modification-related PCD is of significance for AD progression and early prediction, providing insights from a new perspective for the study of disease mechanisms in AD.
Collapse
Affiliation(s)
- Ke Ye
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xinyu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Mengjie Tian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Lulu Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, 150081, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, Heilongjiang, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, 150081, Heilongjiang, China
| | - Qing Xia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, Heilongjiang, China.
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
24
|
Tang M, Dirks K, Kim SY, Qiu Z, Gao Y, Sun D, Peruggia G, Sallavanti J, Li W. Inhibition of thioredoxin reductase 1 sensitizes glucose-starved glioblastoma cells to disulfidptosis. Cell Death Differ 2025; 32:598-612. [PMID: 39715824 PMCID: PMC11982235 DOI: 10.1038/s41418-024-01440-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024] Open
Abstract
Disulfidptosis is a recently identified form of cell death characterized by the aberrant accumulation of cellular disulfides. This process primarily occurs in glucose-starved cells expressing higher levels of SLC7A11 and has been proposed as a therapeutic strategy for cancers with hyperactive SCL7A11. However, the potential for inducing disulfidptosis through other mechanisms in cancers remains unclear. Here, we found that inhibiting thioredoxin reductase 1 (TrxR1), a key enzyme in the thioredoxin system, induces disulfidptosis in glioblastoma (GBM) cells. TrxR1 expression is elevated in GBM with activated transcriptional coactivator with PDZ-binding motif (TAZ) and correlates with poor prognosis. TrxR1 inhibitors induced GBM cell death that can be rescued by disulfide reducers but not by ROS scavengers or inhibitors of apoptosis, ferroptosis, or necroptosis. Glucose-starved cells, but not those deprived of oxygen or glutamine, increased TrxR1 expression in an NRF2-dependent manner and were more sensitive to TrxR1 inhibition-induced cell death. The dying cells initially exhibited highly dynamic lamellipodia, followed by actin cytoskeleton collapse. This process involved the accumulation of cytosolic peroxisomes and micropinocytic caveolae, as well as small gaps in the plasma membrane. Depletion of the WAVE complex component NCKAP1 partially rescued the cells, whereas Rac inhibition enhanced cell death. In an orthotopic xenograft GBM mouse model, TrxR1 depletion inhibited tumor growth and improved survival. Furthermore, cells undergoing TrxR1 inhibition exhibited features of immunogenic cell death. Therefore, this study suggests the potential of targeting TrxR1 as a therapeutic strategy in GBM.
Collapse
Affiliation(s)
- Miaolu Tang
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Kaitlyn Dirks
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
- Colorado State University, Fort Collins, USA
| | - Soo Yeon Kim
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Zhiqiang Qiu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Yan Gao
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Dongxiao Sun
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Gabrielle Peruggia
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Jessica Sallavanti
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA.
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA.
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
25
|
Duan H, Gao L, Asikaer A, Liu L, Huang K, Shen Y. Prognostic Model Construction of Disulfidptosis-Related Genes and Targeted Anticancer Drug Research in Pancreatic Cancer. Mol Biotechnol 2025; 67:1463-1482. [PMID: 38575817 DOI: 10.1007/s12033-024-01131-8] [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: 10/23/2023] [Accepted: 02/19/2024] [Indexed: 04/06/2024]
Abstract
Pancreatic cancer stands as one of the most lethal malignancies, characterized by delayed diagnosis, high mortality rates, limited treatment efficacy, and poor prognosis. Disulfidptosis, a recently unveiled modality of cell demise induced by disulfide stress, has emerged as a critical player intricately associated with the onset and progression of various cancer types. It has emerged as a promising candidate biomarker for cancer diagnosis, prognosis assessment, and treatment strategies. In this study, we have effectively established a prognostic risk model for pancreatic cancer by incorporating multiple differentially expressed long non-coding RNAs (DElncRNAs) closely linked to disulfide-driven cell death. Our investigation delved into the nuanced relationship between the DElncRNA-based predictive model for disulfide-driven cell death and the therapeutic responses to anticancer agents. Our findings illuminate that the high-risk subgroup exhibits heightened susceptibility to the small molecule compound AZD1208, positioning it as a prospective therapeutic agent for pancreatic cancer. Finally, we have elucidated the underlying mechanistic potential of AZD1208 in ameliorating pancreatic cancer through its targeted inhibition of the peroxisome proliferator-activated receptor-γ (PPARG) protein, employing an array of comprehensive analytical methods, including molecular docking and molecular dynamics (MD) simulations. This study explores disulfidptosis-related genes, paving the way for the development of targeted therapies for pancreatic cancer and emphasizing their significance in the field of oncology. Furthermore, through computational biology approaches, the drug AZD1208 was identified as a potential treatment targeting the PPARG protein for pancreatic cancer. This discovery opens new avenues for exploring targets and screening drugs for pancreatic cancer.
Collapse
Affiliation(s)
- Hongtao Duan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Li Gao
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Aiminuer Asikaer
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Lingzhi Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Kuilong Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Yan Shen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China.
| |
Collapse
|
26
|
Wei S, Cui X, Li T, Ma X, Liu L. Pillar[n]arene-Based Supramolecular Nanodrug Delivery Systems for Cancer Therapy. ChemMedChem 2025; 20:e202400822. [PMID: 39833508 DOI: 10.1002/cmdc.202400822] [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: 10/18/2024] [Revised: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Macrocyclic supramolecular materials play an important role in encapsulating anticancer drugs to improve the anticancer efficiency and reduce the toxicity to normal tissues through host-guest interactions. Among them, pillar[n]arenes, as an emerging class of supramolecular macrocyclic compounds, have attracted increasing attention in drug delivery and drug-controlled release due to their high biocompatibility, excellent host-guest chemistry, and simplicity of modification. In this review, we summarize the research progress of pillar[n]arene-based supramolecular nanodrug delivery systems (SNDs) in recent years in the field of tumor therapy, including drug-controlled release, imaging diagnostics and therapeutic modalities. Furthermore, the opportunities and major limitations of pillar[n]arene-based SNDs for tumor therapy are discussed.
Collapse
Affiliation(s)
- Shubin Wei
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
| | - Xinyi Cui
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
| | - Tingting Li
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
| | - Xin Ma
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
| | - Luzhi Liu
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, Guangxi, P. R. China
- Guangxi Key Laboratory of Green Chemical Materials and Safety Technology, Guangxi Engineering Research Center for New Chemical Materials and Safety Technology, College of Petroleum and Chemical Engineering, Beibu Gulf University, Qinzhou, Guangxi, 535011, PR China
| |
Collapse
|
27
|
Yang X, Yang Y, Zhao M, Bai H, Fu C. Identification of DYRK2 and TRIM32 as keloids programmed cell death-related biomarkers: insights from bioinformatics and machine learning in multiple cohorts. Comput Methods Biomech Biomed Engin 2025:1-15. [PMID: 40127455 DOI: 10.1080/10255842.2025.2482129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025]
Abstract
This study aims to explore the expression patterns and mechanisms of programmed cell death-related genes in keloids and identify molecular targets for early diagnosis and treatment. We first explored the expression, immune, and biological function profiles of keloids. Using various machine learning methods, two key genes, DYRK2 and TRIM32, were identified, with ROC curves demonstrating their diagnostic potential. Further analyses, including GSEA, immune cell profiling, competing endogenous RNA network, and single-cell analysis, revealed their mechanism of action and regulatory network. Finally, SB-431542 was identified as a potential therapeutic agent for keloids through CMap and molecular docking.
Collapse
Affiliation(s)
- Xi Yang
- Department of Plastic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yao Yang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Mingjian Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - He Bai
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chongyang Fu
- Department of Hand and Microsurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
28
|
Chang J, Liu D, Xiao Y, Tan B, Deng J, Mei Z, Liao J. Disulfidptosis: a new target for central nervous system disease therapy. Front Neurosci 2025; 19:1514253. [PMID: 40109666 PMCID: PMC11920580 DOI: 10.3389/fnins.2025.1514253] [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: 10/20/2024] [Accepted: 01/27/2025] [Indexed: 03/22/2025] Open
Abstract
Disulfidptosis is a pathologic process that occurs under conditions of NADPH deficiency and excess disulfide bonds in cells that express high levels of SLC7A11. This process is caused by glucose deprivation-induced disulfide stress and was first described by cancer researchers. Oxidative stress is a hypothesized mechanism underlying diseases of the central nervous system (CNS), and disulfide stress is a specific type of oxidative stress. Proteins linked to disulfidptosis and metabolic pathways involved in disulfidptosis are significantly associated with diseases of the CNS (neurodegenerative disease, neurogliomas and ischemic stroke). However, the specific mechanism responsible for this correlation remains unknown. This review provides a comprehensive overview of the current knowledge regarding the origin elements, genetic factors, and signaling proteins involved in the pathogenesis of disulfidptosis. It demonstrates that the disruption of thiometabolism and disulfide stress play critical roles in CNS diseases, which are associated with the potential role of disulfidptosis. We also summarize disulfidptosis-related drugs and highlight potential therapeutic strategies for treating CNS diseases. Additionally, this paper suggests a testable hypothesis that might be a promising target for treating CNS diseases.
Collapse
Affiliation(s)
- Jing Chang
- College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Danhong Liu
- Institute of Clinical Pharmacology of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Chinese Medicine), Changsha, China
| | - Yuqi Xiao
- College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Boyao Tan
- College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Jun Deng
- Department of Neurology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jun Liao
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
- Vascular Biology Laboratory, Medical College, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
29
|
Li S, Li S, Yang D, Zhang J, Wang S, Zeng Z, Cai Q, Zhou Q. NRF2-mediated osteoblast anti-ferroptosis effect promotes induced membrane osteogenesis. Bone 2025; 192:117384. [PMID: 39732449 DOI: 10.1016/j.bone.2024.117384] [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: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Induced membrane technique (IMT) is a new method for repairing segmental bone defects. However, the mechanism of its defect repair is not clear. In recent years, several studies have gradually indicated that ferroptosis is closely related to bone remodeling. Therefore, this study mainly explored the impact of NRF2-mediated osteoblast anti-ferroptosis on bone mineralization within the induced membrane. Male Sprague-Dawley rats aged 12-14 weeks were randomly divided into four groups (n = 12): Model group, DMF (NRF2 agonist) group, ML385 (NRF2 inhibitor) group and Sham group. Except for Sham group, an IMT model of the right femur was established in all other groups. After 4 weeks and 8 weeks of treatment with DMF and ML385, compared to Model group, DMF group showed significantly higher levels of bone volume fraction (BV/TV), osteogenic factors and NRF2/ARE pathway-related factors (NRF2, GPX4, HO-1 and SLC7A11), while ferroptosis-related indicators (total iron, 4-HNE and MDA) were significantly lower. Conversely, ML385 group exhibited significantly higher ferroptosis-related indicators and lower levels of NRF2/ARE pathway-related factors and osteogenesis. In vitro, erastin could induce ferroptosis in osteoblasts. Compared to Erastin group, Erastin+oe-NRF2 (NRF2 overexpression) group showed significantly increased cell viability, mineralization ability, and levels of NRF2/ARE pathway-related factors, along with reduced ferroptosis effects. However, Erastin+si-NRF2 (NRF2 small interfering) group displayed enhanced ferroptosis effects and significantly reduced cell viability, mineralization ability, and levels of NRF2/ARE pathway-related factors. In conclusion, in the bone grafting area of the induced membrane, there existed ferroptosis caused by iron overload. Activating the anti-ferroptosis effect of osteoblasts mediated by the NRF2/ARE signaling cascade could promote growth and mineralization of bone grafts within the induced membrane.
Collapse
Affiliation(s)
- Shuyuan Li
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuying Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medicinal University, Hangzhou 311400, China
| | - Dawen Yang
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingtao Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Songyang Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhanpeng Zeng
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qunbin Cai
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qishi Zhou
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
30
|
Shi M, Li X, Guo Y, Zhang Y, Xu J, Yan L, Liu R, Wang H, Tang S, Zhao Y, Li Z, Feng Y, Ren D, Liu P. Gaudichaudione H Enhances the Sensitivity of Hepatocellular Carcinoma Cells to Disulfidptosis via Regulating NRF2-SLC7A11 Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411131. [PMID: 39840687 PMCID: PMC11923960 DOI: 10.1002/advs.202411131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/01/2024] [Indexed: 01/23/2025]
Abstract
Gaudichaudione H (GH) is a naturally occurring small molecular compound derived from Garcinia oligantha Merr. (Clusiaceae), but the full pharmacological functions remain unclear. Herein, the potential of GH in disulfidptosis regulation, a novel form of programmed cell death induced by disulfide stress is explored. The omics results indicated that NRF2 signaling could be significantly activated by GH. The potential targets are associated with hepatocarcinogenesis and cell death. Moreover, both glutathione (GSH) metabolism and NADP+-NADPH metabolism are affected by GH, indicating the potential in disulfidptosis regulation. It is also observed that GH enhanced the sensitivity of hepatocellular carcinoma (HCC) cells to disulfidptosis, which is dependent on the activation of NRF2-SLC7A11 pathway. GH significantly increased the levels of NRF2 and promoted the transcription of NRF2 target gene, SLC7A11, through autophagy-mediated non-canonical mechanism. Under the condition of glucose starvation, GH-induced upregulation of SLC7A11 aggravated uptake of cysteine, disturbance of GSH synthesis, depletion of NADPH, and accumulation of disulfide molecules, ultimately leading to the formation of disulfide bonds between different cytoskeleton proteins and disulfidptosis eventually. Collectively, the findings underscore the potential role of GH in promoting cancer cell disulfidptosis, thereby offering a promising avenue for the treatment of drug-resistant HCC in clinical settings.
Collapse
Affiliation(s)
- Mengjiao Shi
- Department of General SurgeryNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Xinyan Li
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Ying Guo
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Yinggang Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Jiayi Xu
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Liangwen Yan
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Rongrong Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Hong Wang
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Shenkang Tang
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Department of OncologyAffiliated Hospital of Shaanxi University of Chinese MedicineXianyang712000China
| | - Yaping Zhao
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Zongfang Li
- Department of General SurgeryNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Yetong Feng
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Core Research LaboratoryThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Dongmei Ren
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Key Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical SciencesShandong UniversityJinan250012China
| | - Pengfei Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Key Laboratory of Environment and Genes Related To DiseasesXi'an Jiaotong UniversityMinistry of Education of ChinaXi'an710061China
| |
Collapse
|
31
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
32
|
Jiang Y, Meng H, Zhang X, Yang J, Sun C, Wang X. Identification of subtypes and biomarkers associated with disulfidptosis-related ferroptosis in ulcerative colitis. Hereditas 2025; 162:27. [PMID: 39987439 PMCID: PMC11846262 DOI: 10.1186/s41065-025-00390-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Disulfidptosis and ferroptosis are different programmed cell death modes, which are closely related to the development of a variety of diseases, but the relationship between them and ulcerative colitis (UC) is still unclear. Therefore, our study aimed to explore the molecular subtypes and biomarkers associated with disulfidptosis-related ferroptosis (DRF) in UC. METHODS We used Pearson analysis to identify DRF genes. Then, we classified 140 UC samples into different subtypes based on the DRF genes and explored the biological and clinical characteristics between them. Next, the hub genes were identified by differential analysis and WGCNA algorithms, and three machine learning algorithms were used to screen biomarkers for UC from hub genes. In addition, we analyzed the relationship between biomarkers of immune cells and transcription factors and predicted natural compounds that might be used to treat UC. Finally, we further verified the reliability of the markers by RT-qPCR experiments. RESULTS 118 DRF genes were identified using Pearson analysis. Based on the expression level of the DRF genes, we classified UC patients into C1 and C2 subtypes, with significant differences in the abundance of immune infiltration and disease activity between the two subtypes. The machine learning algorithms identified three biomarkers, including XBP1, FH, and MAP3K5. Further analyses revealed that the three biomarkers were closely associated with a variety of immune cells and transcription factors. In addition, six natural compounds corresponding to the biomarkers were predicted, which may contribute to the effective treatment of UC. Finally, the expression trends of XBP1, FH, and MAP3K5 in animal experiments were consistent with the results of bioinformatics analysis. CONCLUSION In this study, we systematically elucidated the role of DRF genes in the development of UC, and identified three potential biomarkers, providing a new idea for the diagnosis and treatment of UC.
Collapse
Affiliation(s)
- Yinghao Jiang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Hongyan Meng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xin Zhang
- Department of Gastroenterology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jinguang Yang
- Staff Hospital of JIER MACHINE-TOOL GROUP CO.,LTD, Jinan, China
| | - Chengxin Sun
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaoyan Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
33
|
Zhou Z, Zhang Y, Zhou Y, Gu J, Li J, Shao J, Feng N. Construction and verification of a prognostic model for bladder cancer based on disulfidptosis-related angiogenesis genes. PeerJ 2025; 13:e18911. [PMID: 39995998 PMCID: PMC11849515 DOI: 10.7717/peerj.18911] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/06/2025] [Indexed: 02/26/2025] Open
Abstract
Background Bladder cancer (BLCA) is the most common malignancy of the urinary system and one of the most common cancers worldwide. This study seeks to examine the influence of angiogenesis-related genes (ARGs) linked to disulfidptosis on BLCA patients and to formulate a prognostic model for evaluating their prognosis and response to immunotherapy. Methods This study used sequencing data of BLCA in the Cancer Genome Atlas (TCGA) database. Unsupervised consensus clustering analysis, cox regression analysis, and least absolute shrinkage and selection operator (LASSO) regression analysis were used to screen hub genes and construct a related prognostic risk model. The receiver operating characteristic (ROC) curve and independent prognostic analysis were then used to verify the predictive performance of the signature genes. Clinical characteristics, immune status, and Tumor Mutation Burden (TMB) of the prognostic risk model were evaluated. The expression levels of model genes within standard bladder epithelial cell lines (SV-HUC-1) and bladder cancer cell lines (T24 and SW1710) were quantified through qRT-PCR. Results The constructed prognostic risk model can be used as an independent risk indicator for BLCA and was validated in an external dataset. Immune cell infiltration analysis showed that CD8+T cells, Tregs and dendritic cells were significantly different between the two groups. A significant increase was observed in the Stromal score, Immune score and ESTIMATE score in the high-risk group compared with the low-risk group. The Immune Exclusion score and Tumor Immune Dysfunction and Exclusion (TIDE) score of the high-risk group were higher than those of the low-risk score group. Compared with the normal bladder epithelial cell line (SV-HUC-1), the expression levels of 2 model genes (COL5A2 and SCG2) in bladder cancer cell lines (T24 and SW1710) were significantly elevated. Conclusion This study helps us understand the characteristics of disulfidptosis-related subgroups. The characteristics of disulfidptosis-related ARGs may be used to evaluate the prognosis and immunotherapy response of BLCA patients.
Collapse
Affiliation(s)
- Zhihao Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Yuwei Zhang
- Medical School of Nantong University, Nantong, China
| | - Yuhua Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Jiayu Gu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Jufa Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Jianfeng Shao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Ninghan Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| |
Collapse
|
34
|
Xiong X, Zheng LW, Ding Y, Chen YF, Cai YW, Wang LP, Huang L, Liu CC, Shao ZM, Yu KD. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther 2025; 10:49. [PMID: 39966355 PMCID: PMC11836418 DOI: 10.1038/s41392-024-02108-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/27/2024] [Accepted: 12/08/2024] [Indexed: 02/20/2025] Open
Abstract
Breast cancer, characterized by unique epidemiological patterns and significant heterogeneity, remains one of the leading causes of malignancy-related deaths in women. The increasingly nuanced molecular subtypes of breast cancer have enhanced the comprehension and precision treatment of this disease. The mechanisms of tumorigenesis and progression of breast cancer have been central to scientific research, with investigations spanning various perspectives such as tumor stemness, intra-tumoral microbiota, and circadian rhythms. Technological advancements, particularly those integrated with artificial intelligence, have significantly improved the accuracy of breast cancer detection and diagnosis. The emergence of novel therapeutic concepts and drugs represents a paradigm shift towards personalized medicine. Evidence suggests that optimal diagnosis and treatment models tailored to individual patient risk and expected subtypes are crucial, supporting the era of precision oncology for breast cancer. Despite the rapid advancements in oncology and the increasing emphasis on the clinical precision treatment of breast cancer, a comprehensive update and summary of the panoramic knowledge related to this disease are needed. In this review, we provide a thorough overview of the global status of breast cancer, including its epidemiology, risk factors, pathophysiology, and molecular subtyping. Additionally, we elaborate on the latest research into mechanisms contributing to breast cancer progression, emerging treatment strategies, and long-term patient management. This review offers valuable insights into the latest advancements in Breast Cancer Research, thereby facilitating future progress in both basic research and clinical application.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Le-Wei Zheng
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu Ding
- Department of Breast and Thyroid, Guiyang Maternal and Child Health Care Hospital & Guiyang Children's Hospital, Guiyang, P. R. China
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, P. R. China
| | - Yu-Fei Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu-Wen Cai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Lei-Ping Wang
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China.
| |
Collapse
|
35
|
Wang S, Liu X, Tian J, Liu S, Ke L, Zhang S, He H, Shang C, Yang J. Bioinformatics analysis of genes associated with disulfidptosis in spinal cord injury. PLoS One 2025; 20:e0318016. [PMID: 39951434 PMCID: PMC11828381 DOI: 10.1371/journal.pone.0318016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/07/2025] [Indexed: 02/16/2025] Open
Abstract
Research findings indicate that programmed cell death (PCD) plays a pivotal role in the pathophysiology of spinal cord injury (SCI), and a recently discovered form of cell death, disulfidptosis, has emerged as a novel phenomenon. However, the characterization of disulfidptosis-related genes in SCI remains insufficiently explored. We retrieved SCI-related data from the Gene Expression Omnibus (GEO) database and identified three key genes associated with disulfidptosis in human SCI (CAPZB, SLC3A2, and TLN1), whose mediated signaling pathways are closely intertwined with SCI. Subsequent functional enrichment analysis suggested that these genes may regulate multiple pathways and exert corresponding roles in SCI pathology. Moreover, we predicted potential targeted drugs for the key genes along with their transcription factors and constructed an intricate regulatory network. CIBERSORT analysis revealed that CAPZB, SLC3A2, and TLN1 might be implicated in modulating changes within the immune microenvironment of individuals with SCI. Our study provides compelling evidence confirming the significant involvement of disulfidptosis following SCI while offering valuable insights into its underlying pathological mechanisms.
Collapse
Affiliation(s)
- Shuang Wang
- Shangnan County Hospital, Shangnan County, Shangluo City, Shaanxi Province, China
| | - Xinhua Liu
- Shangnan County Hospital, Shangnan County, Shangluo City, Shaanxi Province, China
| | - Jun Tian
- Shangnan County Hospital, Shangnan County, Shangluo City, Shaanxi Province, China
| | - Sizhu Liu
- Shangnan County Hospital, Shangnan County, Shangluo City, Shaanxi Province, China
| | - Lianwei Ke
- Shangnan County Hospital, Shangnan County, Shangluo City, Shaanxi Province, China
| | - Shuling Zhang
- Shangnan County Hospital, Shangnan County, Shangluo City, Shaanxi Province, China
| | - Hongying He
- Shangnan County Hospital, Shangnan County, Shangluo City, Shaanxi Province, China
| | - Chaojiang Shang
- Shangnan County Hospital, Shangnan County, Shangluo City, Shaanxi Province, China
| | - Jichun Yang
- Shangnan County Hospital, Shangnan County, Shangluo City, Shaanxi Province, China
| |
Collapse
|
36
|
Jiang C, Yue T, Jia Z, Song L, Zeng X, Bao Z, Li X, Cui Z, Mi W, Li Q. Disulfidptosis links the pathophysiology of ulcerative colitis and immune infiltration in colon adenocarcinoma. Sci Rep 2025; 15:5365. [PMID: 39948102 PMCID: PMC11825938 DOI: 10.1038/s41598-025-89128-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Ulcerative colitis (UC), a chronic inflammatory bowel disease, significantly increases the risk of colon adenocarcinoma (COAD). Disulfidptosis, a novel form of programmed cell death, has been implicated in various diseases, including UC. This study investigates the expression of disulfidptosis-related genes, particularly CD2AP and MYH10, in UC and COAD. Through analysis of public datasets, we found MYH10 significantly upregulated and CD2AP downregulated in UC compared to healthy controls, with consistent patterns in COAD. Immune infiltration analysis revealed correlations between these genes and specific immune cell types, suggesting their roles in immune modulation. Molecular docking showed strong binding affinities of UC drugs such as budesonide and sulfasalazine with CD2AP and MYH10. Connectivity Map analysis identified additional drug candidates, including simvastatin and mephenytoin, which may be repurposed for UC and COAD therapy. These findings suggest disulfidptosis-related genes as potential biomarkers and therapeutic targets, linking chronic inflammation to cancer progression.
Collapse
Affiliation(s)
- Chenhao Jiang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Teng Yue
- Epidemiology and Biostatistics Institute, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Ziyao Jia
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Lili Song
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaohang Zeng
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ziyu Bao
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xinying Li
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Zhuang Cui
- Epidemiology and Biostatistics Institute, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Wenyi Mi
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Qianqian Li
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
37
|
Guo Z, Liu Y, Chen D, Sun Y, Li D, Meng Y, Zhou Q, Zeng F, Deng G, Chen X. Targeting regulated cell death: Apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis in anticancer immunity. J Transl Int Med 2025; 13:10-32. [PMID: 40115032 PMCID: PMC11921819 DOI: 10.1515/jtim-2025-0004] [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] [Indexed: 03/22/2025] Open
Abstract
In the evolving landscape of cancer treatment, the strategic manipulation of regulated cell death (RCD) pathways has emerged as a crucial component of effective anti-tumor immunity. Evidence suggests that tumor cells undergoing RCD can modify the immunogenicity of the tumor microenvironment (TME), potentially enhancing its ability to suppress cancer progression and metastasis. In this review, we first explore the mechanisms of apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis, along with the crosstalk between these cell death modalities. We then discuss how these processes activate antigen-presenting cells, facilitate the cross-priming of CD8+ T cells, and trigger anti-tumor immune responses, highlighting the complex effects of novel forms of tumor cell death on TME and tumor biology. Furthermore, we summarize potential drugs and nanoparticles that can induce or inhibit these emerging RCD pathways and their therapeutic roles in cancer treatment. Finally, we put forward existing challenges and future prospects for targeting RCD in anti-cancer immunity. Overall, this review enhances our understanding of the molecular mechanisms and biological impacts of RCD-based therapies, providing new perspectives and strategies for cancer treatment.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yihuang Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Danyao Chen
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yuming Sun
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Furong Zeng
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
38
|
Yao HF, Ge J, Chen J, Tang X, Li C, Hu X, Ahmed AA, Pu Y, Zhou G, Zhang T, Cai Z, Jiang C. CASC8 activates the pentose phosphate pathway to inhibit disulfidptosis in pancreatic ductal adenocarcinoma though the c-Myc-GLUT1 axis. J Exp Clin Cancer Res 2025; 44:26. [PMID: 39865281 PMCID: PMC11771065 DOI: 10.1186/s13046-025-03295-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 01/15/2025] [Indexed: 01/28/2025] Open
Abstract
PURPOSE Glucose starvation induces the accumulation of disulfides and F-actin collapse in cells with high expression of SLC7A11, a phenomenon termed disulfidptosis. This study aimed to confirm the existence of disulfidptosis in pancreatic ductal adenocarcinoma (PDAC) and elucidate the role of Cancer Susceptibility 8 (CASC8) in this process. METHODS The existence of disulfidptosis in PDAC was assessed using flow cytometry and F-actin staining. CASC8 expression and its clinical correlations were analyzed using data from The Cancer Genome Atlas (TCGA) and further verified by chromogenic in situ hybridization assay in PDAC tissues. Cells with CASC8 knockdown and overexpression were subjected to cell viability, EdU, transwell assays, and used to establish subcutaneous and orthotopic tumor models. Disulfidptosis was detected by flow cytometry and immunofluorescence assays. RNA sequencing and metabolomics analysis were performed to determine the metabolic pathways which were significantly affected after CASC8 knockdown. We detected the glucose consumption and the NADP+/NADPH ratio to investigate alterations in metabolic profiles. RNA immunoprecipitation combined with fluorescence in situ hybridization assay was used to identify protein-RNA interactions. Protein stability, western blotting and quantitative real-time PCR assays were performed to reveal potential molecular mechanism. RESULTS Disulfidptosis was observed in PDAC and could be significantly rescued by disulfidptosis inhibitors. CASC8 expression was higher in PDAC samples compared to normal pancreatic tissue. High CASC8 expression correlated with a poor prognosis for patients with PDAC and contributed to cancer progression in vitro and in vivo. Furthermore, CASC8 was associated with disulfidptosis resistance under glucose starvation conditions in PDAC. Mechanistically, CASC8 interacted with c-Myc to enhance the stability of c-Myc protein, leading to the activation of the pentose phosphate pathway, a reduction of the NADP+/NADPH ratio and ultimately inhibiting disulfidptosis under glucose starvation conditions. CONCLUSIONS This study provides evidence for the existence of disulfidptosis in PDAC and reveals the upregulation of CASC8 in this malignancy. Furthermore, we demonstrate that CASC8 acts as a crucial regulator of the pentose phosphate pathway and disulfidptosis, thereby promoting PDAC progression.
Collapse
Affiliation(s)
- Hong-Fei Yao
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Jieqiong Ge
- Department of Nursing, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Jiahao Chen
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Xiaoyan Tang
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Chunjing Li
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Xiao Hu
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Abousalam Abdoulkader Ahmed
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Yunlong Pu
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Guihua Zhou
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Tongyi Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Zhiwei Cai
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China.
| | - Chongyi Jiang
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, PR China.
| |
Collapse
|
39
|
Zhang X, Lin Y, Shi L, Zhai A, Wu C, Zhu QY. Disulfidptosis-related gene SLC3A2: a novel prognostic biomarker in nasopharyngeal carcinoma and head and neck squamous cell carcinoma. Front Oncol 2025; 15:1451034. [PMID: 39926285 PMCID: PMC11802814 DOI: 10.3389/fonc.2025.1451034] [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: 06/18/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Introduction Nasopharyngeal carcinoma (NPC), one of the most common malignancies of the head and neck, is characterised by a complex pathogenesis and an unfavourable prognosis. Recently, disulfidoptosis, a novel form of cell death, has been proposed. Several studies in recent years have extensively investigated the function of the disulfidoptosis-related SLC7A11 gene in cancer, but the role of its partner protein, SLC3A2, remains unknown unclear in NPC. Methods GEO database analysis confirmed SLC3A2's prognostic impact on nasopharyngeal carcinoma. ROC, Kaplan-Meier analyses, and stage-specific expression studies showed a strong correlation with poor HNSC prognosis. GO and KEGG analyses pinpointed relevant signaling pathways. In vitro, SLC3A2's influence on cell proliferation, migration, and invasion was evaluated through CCK8, wound healing, colony formation, transwell assays, and cell cycle analysis. Results In this study, we identified the high expression of SLC3A2 in NPC and head and neck squamous cell carcinoma (HNSC) and analyzed its potential mechanism and correlation with patient prognosis. Furthermore, a negative relationship was found between the expression level of SLC3A2 and the extent of immune cell infiltration and immune checkpoint. Differentially expressed genes (DEGs) between the high and low SLC3A2 expression groups were primarily involved in cytokine-cytokine receptor interaction and immune response. Finally, in vitro experiments demonstrated that SLC3A2 stimulates tumor cell proliferation and migration. Discussion In conclusion, these results indicated a strong association between SLC3A2 and progression in both NPC and HNSC, suggesting it as a promising biomarker for predicting adverse prognosis in NPC and HNSC patients.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yiqi Lin
- Department of Endocrinology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Liang Shi
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Aixia Zhai
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chao Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Qian-Ying Zhu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
40
|
Zhou Y, Zhang Y, Zhou Y, Gu Y, Chen Y, Wang J. Bioinformation study of immune microenvironment characteristics of disulfidptosis-related subtypes in ovarian cancer and prognostic model construction. Discov Oncol 2025; 16:18. [PMID: 39775403 PMCID: PMC11711411 DOI: 10.1007/s12672-025-01752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE Ovarian cancer significantly impacts women's reproductive health and remains challenging to diagnose and treat. Despite advancements in understanding DNA repair mechanisms and identifying novel therapeutic targets, additional strategies are still needed. Recently, a novel form of cell death called disulfidptosis, which is triggered by glucose deprivation, has been linked to treatment resistance and changes in the tumor microenvironment (TME). However, its role in ovarian cancer is not well understood. METHODS Bioinformatics analysis was performed on RNA-seq data from TCGA and GEO databases to identify disulfidptosis-related genes in ovarian cancer. Differential expression analysis and pathway enrichment were conducted, followed by the development of a prognostic model using LASSO Cox regression, validated with GEO datasets (GSE13876, GSE26712). Clinical samples were analyzed using quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC) to validate gene expression. RESULTS This study identified disulfidptosis-related gene subtypes in ovarian cancer and demonstrated their influence on the tumor microenvironment (TME), immunotherapy responses, and patient prognosis. Six genes (IFNB1, IGF2, CD40LG, IL1B, IL21, CD38) associated with disulfidptosis were identified and incorporated into a prognostic model. This model predicted patient outcomes and was validated externally. Clinical validation showed its accuracy in predicting progression-free survival and resistance to platinum-based chemotherapy. CONCLUSION Our findings highlight the significant impact of disulfidptosis-related genes on the ovarian cancer tumor microenvironment, providing insights that could support the development of clinical evaluations and personalized treatment strategies.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yuhong Zhang
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, Suzhou, Jiangsu, China
| | - Yang Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanzheng Gu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Juan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
41
|
Wei M, Shi X, Tang W, Lv Q, Wu Y, Xu Y. Identification of a novel disulfidptosis-related gene signature in osteoarthritis using bioinformatics analysis and experimental validation. Sci Rep 2025; 15:1339. [PMID: 39779817 PMCID: PMC11711464 DOI: 10.1038/s41598-025-85569-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: 04/26/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative bone disease characterized by the destruction of joint cartilage and synovial inflammation, involving intricate immune regulation processes. Disulfidptosis, a novel form of programmed cell death, has recently been identified; however, the effects and roles of disulfidptosis-related genes (DR-DEGs) in OA remain unclear. We obtained six OA datasets from the GEO database, using four as training sets and two as validation sets. Differential expression analysis was employed to identify DR-DEGs, and unique molecular subtypes of OA were constructed based on these DR-DEGs. Subsequently, the immune microenvironment of OA patients was comprehensively analyzed using the "CIBERSORT" algorithm for immune infiltration. Various machine learning algorithms were utilized to screen characteristic DR-DEGs, and nomogram models and ROC curves were built based on these genes. The scRNA dataset (GSE169454) was used to classify chondrocytes in OA samples into distinct cell types, further exploring the gene distribution and correlation of characteristic DR-DEGs with specific cell subpopulations. Moreover, the expression levels of four characteristic DR-DEGs were validated through OA cell models and rat models. In our study, we identified 10 DR-DEGs with significant differences in expression within OA samples. Based on these DR-DEGs, two distinct molecular subtypes were recognized (cluster 1 and 2). ZNF484 and NDUFS1 were found to be significantly overexpressed in subtype 1, while the infiltration abundance of activated mast cells was markedly elevated in subtype 2. Moreover, significant differences were observed in the infiltration proportions of 11 immune cell types between OA and control samples, with 9 DR-DEGs demonstrating substantial correlations with immune cell infiltration levels. Further analysis of the scRNA dataset revealed that SLC3A2 and NDUFC1 were predominantly expressed in the preHTC subpopulation. All 10 DR-DEGs exhibited notably higher expression in the EC subpopulation across various cell types. The proportion of EC subgroups with high SLC3A2 expression increased, mainly enriching pathways related to inflammation, such as the IL-17 signaling pathway and TGF-beta signaling pathway. Using machine learning, we identified four characteristic DR-DEGs, which, in combination with the nomogram and ROC models, demonstrated promising performance in the diagnosis of OA. Additionally, in vivo validation confirmed a significant elevation of PPM1F expression in OA models. This study identified DR-DEGs as potential biomarkers for the diagnosis and classification of OA and provided a preliminary understanding of their role in the immune microenvironment. However, further experimental and clinical studies are required to validate their diagnostic value and therapeutic potential.
Collapse
Affiliation(s)
- Mingjie Wei
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, China
| | - Xiangwen Shi
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, China
| | - Wenbao Tang
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, China
| | - Qian Lv
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, China
| | - Yipeng Wu
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, China
| | - Yongqing Xu
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, China.
| |
Collapse
|
42
|
Li X, Zhu D. Role of disulfide death in cancer (Review). Oncol Lett 2025; 29:55. [PMID: 39606569 PMCID: PMC11600708 DOI: 10.3892/ol.2024.14801] [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: 07/24/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
The research field of regulated cell death is growing extensively. Following the recognition of ferroptosis, other unique and distinct forms of regulated cell death, including cuproptosis and disulfide death, have been identified. Disulfide death occurs due to the abnormal accumulation of disulfides within cells in environments lacking glucose, leading to contraction of the actin cytoskeleton, which ultimately triggers various signaling pathways and cell death. The induction of disulfide death in the treatment of cancer may exhibit significant therapeutic potential. Therefore, in the present review, a comprehensive and critical analysis of the current understanding of the molecular mechanisms and regulatory networks of disulfide death is presented. In addition, the potential physiological functions of disulfide death in tumor suppression and immune surveillance as well as its pathological roles and therapeutic potential are described. The core focus areas for future research into this form of cell death are also explored. Given the current lack of extensive clinical findings and well-defined key concepts, these may be regarded as pivotal points of interest in future studies.
Collapse
Affiliation(s)
- Xue Li
- Oncology Department, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Danxia Zhu
- Oncology Department, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
43
|
Li Y, Zhang H, Yang F, Zhu D, Chen S, Wang Z, Wei Z, Yang Z, Jia J, Zhang Y, Wang D, Ma M, Kang X. Mechanisms and therapeutic potential of disulphidptosis in cancer. Cell Prolif 2025; 58:e13752. [PMID: 39354653 DOI: 10.1111/cpr.13752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
SLC7A11 plays a pivotal role in tumour development by facilitating cystine import to enhance glutathione synthesis and counteract oxidative stress. Disulphidptosis, an emerging form of cell death observed in cells with high expression of SLC7A11 under glucose deprivation, is regulated through reduction-oxidation reactions and disulphide bond formation. This process leads to contraction and collapse of the F-actin cytoskeleton from the plasma membrane, ultimately resulting in cellular demise. Compared to other forms of cell death, disulphidptosis exhibits distinctive characteristics and regulatory mechanisms. This mechanism provides novel insights and innovative strategies for cancer treatment while also inspiring potential therapeutic approaches for other diseases. Our review focuses on elucidating the molecular mechanism underlying disulphidptosis and its connection with the actin cytoskeleton, identifying alternative metabolic forms of cell death, as well as offering insights into disulphidptosis-based cancer therapy. A comprehensive understanding of disulphidptosis will contribute to our knowledge about fundamental cellular homeostasis and facilitate the development of groundbreaking therapies for disease treatment.
Collapse
Affiliation(s)
- Yanhu Li
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Haijun Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
- The Second People's Hospital of Gansu Province, Lanzhou, PR China
| | - Fengguang Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Daxue Zhu
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Shijie Chen
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhaoheng Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Ziyan Wei
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhili Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Jingwen Jia
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Yizhi Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Dongxin Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Mingdong Ma
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Xuewen Kang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| |
Collapse
|
44
|
Gu X, Wei Y, Lu M, Shen D, Wu X, Huang J. Systematic Analysis of Disulfidptosis-Related lncRNAs in Hepatocellular Carcinoma with Vascular Invasion Revealed That AC131009.1 Can Promote HCC Invasion and Metastasis through Epithelial-Mesenchymal Transition. ACS OMEGA 2024; 9:49986-49999. [PMID: 39713637 PMCID: PMC11656384 DOI: 10.1021/acsomega.4c09411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024]
Abstract
Disulfidptosis, a recently identified pathway of cellular demise, served as the focal point of this research, aiming to pinpoint relevant lncRNAs that differentiate between hepatocellular carcinoma (HCC) with and without vascular invasion while also forecasting survival rates and responses to immunotherapy in patients with vascular invasion (VI+). First, we identified 300 DRLRs in the TCGA database. Subsequently, utilizing univariate analysis, LASSO-Cox proportional hazards modeling, and multivariate analytical approaches, we selected three DRLRs (AC009779.2, AC131009.1, and LUCAT1) with the highest prognostic value to construct a prognostic risk model for VI+ HCC patients. Multivariate Cox regression analysis revealed that this model is an independent prognostic factor for predicting overall survival that outperforms traditional clinicopathological factors. Pathway analysis demonstrated the enrichment of tumor and immune-related pathways in the high-risk group. Immune landscape analysis revealed that immune cell infiltration degrees and immune functions had significant differences. Additionally, we identified valuable chemical drugs (AZD4547, BMS-536924, BPD-00008900, dasatinib, and YK-4-279) for high-risk VI+ HCC patients. In-depth bioinformatics analysis was subsequently conducted to assess immune characteristics, drug susceptibility, and potential biological pathways involving the three hub DRLRs. Furthermore, the abnormally elevated transcriptional levels of the three DRLRs in HCC cell lines were validated through qRT-PCR. Functional cell assays revealed that silencing the expression of lncRNA AC131009.1 can inhibit the migratory and invasive capabilities of HCC cells, a finding further corroborated by the chorioallantoic membrane (CAM) assay. Immunohistochemical analysis and hematoxylin-eosin staining (HE) staining provided preliminary evidence that AC131009.1 may promote the invasion and metastasis of HCC cells by inducing epithelial-mesenchymal transition (EMT) in both subcutaneous xenograft models and orthotopic HCC models within nude mice. To summarize, we developed a risk assessment model founded on DRLRs and explored the potential mechanisms by which hub DRLRs promote HCC invasion and metastasis.
Collapse
Affiliation(s)
- Xuefeng Gu
- Department
of Infectious Diseases, Jurong Hospital
Affiliated to Jiangsu University, Zhenjiang, Jiangsu 212400, China
| | - Yanyan Wei
- Department
of Infectious Diseases, The First Affiliated
Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Mao Lu
- Department
of Gastroenterology, The Affiliated Changzhou
Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Duo Shen
- Department
of Gastroenterology, The Affiliated Changzhou
Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Xin Wu
- Department
of General Surgery, The Fourth Affiliated
Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Jin Huang
- Department
of Gastroenterology, The Affiliated Changzhou
Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, China
| |
Collapse
|
45
|
Han L, Yang H, Jiang X, Zhou Z, Ge C, Yu K, Li G, Wang W, Liu Y. Prognostic model based on disulfidptosis-related lncRNAs for predicting survival and therapeutic response in bladder cancer. Front Immunol 2024; 15:1512203. [PMID: 39687628 PMCID: PMC11647029 DOI: 10.3389/fimmu.2024.1512203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Background With poor treatment outcomes and prognosis, bladder cancer remains a focus for clinical research in the precision oncology era. However, the potential of disulfidptosis, a novel cell death mechanism, and its related long non-coding RNAs to support selective cancer cell killing in this disease is still unclear. Methods We identified key disulfidptosis-related lncRNAs in bladder cancer, constructed a prognostic risk model with potential therapeutic targets, and confirmed the findings through quantitative PCR analysis. Results We identified five crucial lncRNAs (AC005840.4, AC010331.1, AL021707.6, MIR4435-2HG and ARHGAP5-AS1) and integrated them into a predictive model centered on disulfidptosis-associated lncRNAs. Reliability and validity tests demonstrated that the lncRNA prediction index associated with disulfidptosis effectively discerns patients' prognosis outcomes. Additionally, high-risk patients exhibited elevated expression levels of genes involved in the PI3K-Akt signaling pathway, extracellular matrix organization, and immune escape mechanisms, which are associated with poor prognosis. Notably, high-risk patients demonstrated higher sensitivity to Sorafenib, Oxaliplatin and MK-2206, underscoring the promise of these lncRNAs as precise therapeutic targets in bladder cancer. Conclusion By revealing the predictive importance of disulfidptosis-associated lncRNAs in bladder cancer, our research offers new perspectives and pinpoints potential therapeutic targets in clinical environments.
Collapse
Affiliation(s)
- Lirui Han
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Hankai Yang
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Xuan Jiang
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Ziyu Zhou
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Chang Ge
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Kairan Yu
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Guofang Li
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Wei Wang
- Ministry of Education (MOE) Key Laboratory of Bio-Intelligent Manufacturing, Dalian University of Technology, Dalian, China
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Liaoning, Shenyang, China
| | - Yubo Liu
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
- Ministry of Education (MOE) Key Laboratory of Bio-Intelligent Manufacturing, Dalian University of Technology, Dalian, China
| |
Collapse
|
46
|
Xu X, Zhang B, Zhang J, Ma H. Unraveling disulfidptosis for prognostic modeling and personalized treatment strategies in lung adenocarcinoma. Future Sci OA 2024; 10:2432211. [PMID: 39587729 PMCID: PMC11601057 DOI: 10.1080/20565623.2024.2432211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
AIM To construct and identify a prognostic and therapeutic signature based on disulfidptosis-related genes in lung adenocarcinoma. METHODS Bioinformatic analysis was performed to assess the differential expression of disulfidptosis-related genes between cancerous and control samples from The Cancer Genome Atlas-Lung Adenocarcinoma (TCGA-LUAD) database. Survival analysis, immune cell infiltration assessment, and examination of oncogenic pathways were performed to uncover potential clinical implications of disulfidptosis gene expression. Differential gene expression analysis between subtypes facilitated the development of a prognostic model using a combination of genes associated with survival. A nomogram was further created using independent clinical and molecular factors. RESULTS We identified the significant upregulation of ten disulfidptosis-related genes and delineated two distinct subtypes, C1 and C2. Subtype C2 was associated with prolonged survival. Then, prognostic modeling utilizing six genes (TXNRD1, CPS1, S100P, SCGB3A1, CYP24A1, NAPSA) demonstrated predictive power in both training and validation datasets. The nomogram, incorporating the risk model with clinical features, provided a reliable tool for predicting one-year (AUC 0.77), three-year (AUC 0.75), and five-year (AUC 0.78) survival rates. Additionally, chemotherapy sensitivity analysis highlighted significant resistance in the high-risk group, primarily associated with subtype C1. CONCLUSION Our study reveals distinct LUAD subtypes, offers a robust prognostic model, and underscores clinical implications for personalized therapy based on disulfidptosis-related genes expression profiles.
Collapse
Affiliation(s)
- Xiangyu Xu
- Department of Thoracic Surgery, Chongqing General Hospital, Chongqing, China
| | - Bingbing Zhang
- Department of Thoracic Surgery, Chongqing General Hospital, Chongqing, China
| | - Jin Zhang
- Department of Thoracic Surgery, Chongqing General Hospital, Chongqing, China
| | - Hongbiao Ma
- Department of Thoracic Surgery, Chongqing General Hospital, Chongqing, China
| |
Collapse
|
47
|
Chen Q, Zhang C, Meng T, Yang K, Hu Q, Tong Z, Wang X. Prediction of clinical prognosis and drug sensitivity in hepatocellular carcinoma through the combination of multiple cell death pathways. Cell Biol Int 2024; 48:1816-1835. [PMID: 39192561 DOI: 10.1002/cbin.12235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/29/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024]
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignant tumor, highlighting a significant need for reliable predictive models to assess clinical prognosis, disease progression, and drug sensitivity. Recent studies have highlighted the critical role of various programmed cell death pathways, including apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, entotic cell death, NETotic cell death, parthanatos, lysosome-dependent cell death, autophagy-dependent cell death, alkaliptosis, oxeiptosis, and disulfidptosis, in tumor development. Therefore, by investigating these pathways, we aimed to develop a predictive model for HCC prognosis and drug sensitivity. We analyzed transcriptome, single-cell transcriptome, genomic, and clinical information using data from the TCGA-LIHC, GSE14520, GSE45436, and GSE166635 datasets. Machine learning algorithms were used to establish a cell death index (CDI) with seven gene signatures, which was validated across three independent datasets, showing that high CDI correlates with poorer prognosis. Unsupervised clustering revealed three molecular subtypes of HCC with distinct biological processes. Furthermore, a nomogram integrating CDI and clinical information demonstrated good predictive performance. CDI was associated with immune checkpoint genes and tumor microenvironment components using single-cell transcriptome analysis. Drug sensitivity analysis indicated that patients with high CDI may be resistant to oxaliplatin and cisplatin but sensitive to axitinib and sorafenib. In summary, our model offers a precise prediction of clinical outcomes and drug sensitivity for patients with HCC, providing valuable insights for personalized treatment strategies.
Collapse
Affiliation(s)
- QingKun Chen
- Department of Graduate School, Bengbu Medical University, Bengbu, China
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - ChenGuang Zhang
- Department of Graduate School, Bengbu Medical University, Bengbu, China
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - Tao Meng
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - Ke Yang
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - QiLi Hu
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - Zhong Tong
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - XiaoGang Wang
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| |
Collapse
|
48
|
Fang K, Jiang S, Xu Z, Luo M, Yan C. Pan-cancer landscape of disulfidptosis across human tumors. Heliyon 2024; 10:e40122. [PMID: 39605832 PMCID: PMC11600026 DOI: 10.1016/j.heliyon.2024.e40122] [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: 03/27/2024] [Revised: 09/20/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Objective Disulfidptosis is a newly discovered disulfide stress-induced cell death form. Clinical significance and biological mechanisms of disulfidptosis in human cancers need to be further elucidated. Thus, this study was designed to characterize pan-cancer landscape of disulfidptosis across human tumors. Methods Multi-omics features (transcriptomics, genomics, and DNA methylation) of disulfidptosis genes were investigated in TCGA pan-cancer cohorts. A disulfidptosis score system was defined across human tumors via ssGSEA. The activity of classical oncogenic pathways and hallmarks of cancer as well as the infiltration of immunocyte subpopulations were estimated, respectively. Drug sensitivity was inferred, and immune checkpoint blockade (ICB) response was evaluated in an independent cohort IMvigor210. ACHN, CAL-27, and NCI-H23 cells were transiently transfected with GYS1 siRNAs, and cell apoptosis and proliferation were measured through TUNEL and EdU assays, respectively. Results Aberrant mRNA expression and DNA methylation of disulfidptosis genes as well as their genomic alterations were found in human tumors. The disulfidptosis score was utilized for quantifying the activity of disulfidptosis, which enabled to estimate patient prognosis. The disulfidptosis score presented positive correlations to angiogenesis and EMT, indicating the role of disulfidptosis in mediating tumor malignant features. Moreover, the score was negatively linked with infiltrating immune and stromal cells in the immune microenvironment. In the ICB cohort, shorter survival time was observed in patients with high disulfidptosis score, indicating the potential of disulfidptosis score in influencing clinical benefits from ICB. Additionally, tumors with low disulfidptosis score exhibited higher sensitivity to a few small molecular compounds, e.g., Sabutoclax, PRIMA-1MET, BIBR-1532, and Elephantin. Knockdown of disulfidptosis gene GYS1 effectively hindered tumor progression. Conclusion Collectively, our findings depict a pan-cancer map of disulfidptosis to inform functional and therapeutic research.
Collapse
Affiliation(s)
- Kun Fang
- Department of Surgery, Yinchuan Maternal and Child Health Hospital, Yinchuan, 750001, China
| | - Suxiao Jiang
- Department of Surgery, Yinchuan Maternal and Child Health Hospital, Yinchuan, 750001, China
| | - Zhengjie Xu
- Department of Surgery, Yinchuan Maternal and Child Health Hospital, Yinchuan, 750001, China
| | - Meng Luo
- Department of Surgery, Yinchuan Maternal and Child Health Hospital, Yinchuan, 750001, China
| | - Changsheng Yan
- Department of Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| |
Collapse
|
49
|
Wang B, Wang W, Wang Y, Wen X, Wang Z, Leng H, Kong F, Ma X. Analysis and experimental validation of disulfidptosis related genes solute carrier family 3 member 2 (SLC3A2) in endometrial cancer. Cancer Cell Int 2024; 24:390. [PMID: 39609847 PMCID: PMC11606104 DOI: 10.1186/s12935-024-03560-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024] Open
Abstract
Disulfidptosis, a novel cell death paradigm triggered by disulfide stress, remains underexplored, particularly its implications for endometrial cancer (EC). This study focused on the prognostic significance of disulfidptosis-related genes (DRGs) in EC, highlighting the pivotal role of SLC3A2. To predict EC patient outcomes, we developed a model centered on DRGs, employing LASSO-Cox regression for its construction. The model revealed a strong correlation between DRG risk score, gene set enrichment analysis (GSEA), single-sample GSEA (ssGSEA), clinical characteristics, the tumor microenvironment (TME), and the response to immunotherapy. Key genes were pinpointed using random forest maps. To establish SLC3A2's oncogenic effects in EC, we conducted comprehensive studies including apoptosis, cell cycle, TRANSWELL, CCK-8, and tumor xenograft assays. SLC3A2 expression was further confirmed via qRT-PCR. The impact of SLC3A2 on EC's malignant behavior was corroborated through both in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Bo Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No 39 Huaxiang Road, Tiexi District, Shenyang, 110000, Liaoning, People's Republic of China
| | - Wantong Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No 39 Huaxiang Road, Tiexi District, Shenyang, 110000, Liaoning, People's Republic of China
| | - Yuting Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No 39 Huaxiang Road, Tiexi District, Shenyang, 110000, Liaoning, People's Republic of China
| | - Xin Wen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No 39 Huaxiang Road, Tiexi District, Shenyang, 110000, Liaoning, People's Republic of China
| | - Zihao Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No 39 Huaxiang Road, Tiexi District, Shenyang, 110000, Liaoning, People's Republic of China
| | - Hongrui Leng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No 39 Huaxiang Road, Tiexi District, Shenyang, 110000, Liaoning, People's Republic of China
| | - Fanfei Kong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No 39 Huaxiang Road, Tiexi District, Shenyang, 110000, Liaoning, People's Republic of China.
| | - Xiaoxin Ma
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No 39 Huaxiang Road, Tiexi District, Shenyang, 110000, Liaoning, People's Republic of China.
| |
Collapse
|
50
|
Wang Z, Yang J, He P, Lan J, Shi T, Xu S, Hao Z, Xi Y, Wang J, He P. Therapeutic effect of total glucosides of paeony on IgA vasculitis nephritis: progress and prospects. Mol Biol Rep 2024; 52:13. [PMID: 39585482 PMCID: PMC11588768 DOI: 10.1007/s11033-024-10041-6] [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: 07/31/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
IgA vasculitis nephritis (IgAVN), a type of immune system disease characterized by the deposition of IgA in the mesangial area of the glomerulus, is most common in children. Corticosteroids and immunosuppressants agents are commonly prescribed for the clinical management of IgAVN; however, these therapies are associated with numerous adverse reactions. This necessitates the discovery of alternative, potential therapeutic agents that can offer a safer treatment option. Natural plants contain abundant total glucosides of paeony (TGP), which represent one of the most prevalent secondary metabolites found within these organisms. TGP has been proven to be a safe and desirable natural medicine, with the most central ingredient being polyphenolic. TGP, a naturally occurring and cost-effective compound, exerts its therapeutic influence on IgAVN via diverse pathways and targets, with minimal side effects. Its substantial clinical potential underscores the importance of delving deeper into its pharmacological mechanisms, which hold great promise for novel drug development. This review examines the multifaceted therapeutic mechanisms of TGP in IgAVN encompassing modulation of Wnt/β-catenin pathways and programmed cell death pathways, antioxidant and anti-inflammatory effects, and enhancement of vascular repair. Additionally, we provide an overview of recent advancements in enhancing the bioavailability of TGP and highlight crucial considerations that may inform future research endeavors.
Collapse
Affiliation(s)
- Zhifeng Wang
- The First Clinical College of Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jiao Yang
- The Second Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Pengfen He
- The First Clinical College of Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Junfeng Lan
- The First Clinical College of Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Ting Shi
- The First Clinical College of Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Shuangfeng Xu
- The First Clinical College of Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhihui Hao
- The First Clinical College of Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yujiang Xi
- The First Clinical College of Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
| | - Jian Wang
- The First Clinical College of Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
| | - Ping He
- Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China.
| |
Collapse
|