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
|
Zhou QM, Lu YF, Yang XY, Zhang JG, Wang YN, Luo WP, Mao J, Hou J, Wu F, Wang WL, Tang GP, Bai HZ, Yu RS. Redox-driven hybrid nanoenzyme dynamically activating ferroptosis and disulfidptosis for hepatocellular carcinoma theranostics. J Colloid Interface Sci 2025; 693:137611. [PMID: 40253866 DOI: 10.1016/j.jcis.2025.137611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Hepatocellular carcinoma (HCC) presents formidable therapeutic challenges due to its pronounced metabolic heterogeneity, particularly arising from spatially uneven glucose availability within the tumor microenvironment (TME). To address this, we developed a glutathione (GSH)-responsive, biomimetic hybrid nanoenzyme system (M@GOx/Fe-HMON) composed of hollow mesoporous organosilica nanoparticles co-loaded with glucose oxidase (GOx) and Fe2+/Fe3+ redox pairs, and cloaked in homologous tumor cell membranes for enhanced targeting. In glucose-rich regions, the nanoenzyme orchestrates a GOx-peroxidase (POD) cascade that produces reactive oxygen species (ROS) via the Fenton reaction, leading to ferroptosis through intensified oxidative stress and GSH depletion. Conversely, under glucose-deficient conditions, the nanoenzyme promotes disulfidptosis by aggravating glucose deprivation, depleting nicotinamide adenine dinucleotide phosphate (NADPH), and impairing cystine metabolism, ultimately resulting in actin cytoskeletal collapse. This dual-action platform dynamically adapts to the tumor's metabolic landscape, selectively inducing ferroptosis or disulfidptosis according to glucose levels, disrupting redox homeostasis and amplifying antitumor efficacy. Notably, this study is the first to integrate ferroptosis and disulfidptosis activation into a single, metabolism-sensitive nanoenzyme system, providing a novel paradigm for exploiting tumor metabolic heterogeneity. Furthermore, the combination of endogenous metabolic regulation with magnetic resonance imaging (MRI)-guided diagnosis introduces an innovative and noninvasive strategy for precision cancer theranostics.
Collapse
Affiliation(s)
- Qiao-Mei Zhou
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Yuan-Fei Lu
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Xiao-Yan Yang
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Jin-Guo Zhang
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Yi-Ning Wang
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Wang-Ping Luo
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Jin Mao
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Jue Hou
- Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China
| | - Fan Wu
- Department of Neurosurgery, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Wei-Lin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Gu-Ping Tang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China.
| | - Hong-Zhen Bai
- Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China.
| | - Ri-Sheng Yu
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Yao Y, Yang X, Fu Y, Zhang Y. Immunological features of various molecular subtypes of cervical cancer and their prognostic implications in the context of disulfidptosis. Front Oncol 2025; 15:1574911. [PMID: 40438679 PMCID: PMC12116334 DOI: 10.3389/fonc.2025.1574911] [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: 02/11/2025] [Accepted: 04/25/2025] [Indexed: 06/01/2025] Open
Abstract
Objective Cervical cancer ranks among the most prevalent malignancies impacting women globally. Disulfidptosis represents a recently identified pathway of cellular demise, although its role in the context of cervical cancer is not well elucidated. This research investigates the significance of Disulfidptosis-Related Genes (DRGs) within cervical cancer. Furthermore, it aims to analyze the differences in prognosis and immune infiltration among different molecular subtypes. Methods We compiled genes associated with cervical cancer and disulfidptosis from a variety of databases to perform a differential expression analysis. Subsequently, the samples are grouped through consensus clustering. To evaluate immune cell infiltration, we employed CIBERSORT. Additionally, immune checkpoint genes (ICGs) were gathered from existing literature and databases, enabling statistical analyses of two subtype samples of cervical cancer (CESC). Following our analyses using GO, KEGG, and GSEA to compare the differences between the two subtypes. Lastly, a prognostic risk model was constructed using LASSO regression and validated using ROC. Results This study identified seven key genes: PCBP3, ARNT, ANP32E, DSTN, CD2AP, EPAS1, and ACTN1.The consensus clustering analysis showed differences in immune cell infiltration and DFS(disease-free survival) among the various clusters. The immune checkpoint gene CXCL1 displayed highly significant statistical differences between subtype A (Cluster 1) and subtype B (Cluster 2) in cervical cancer (CESC) samples. The gene set enrichment analysis identified the negative regulation of peptidase activity and the IL-17 signaling pathway, which link to subtype-specific differentially expressed genes (DEGs). Conclusion Statistical analysis of the various subtypes of CESC samples highlighted the importance of subtype-specific therapeutic targets. Additionally, it seeks to enhance the accuracy of prognostic predictions, thereby establishing a foundation for the formulation of personalized treatment approaches.
Collapse
Affiliation(s)
- Yadan Yao
- Department of Gynecology, Jiaxing University Affiliated Traditional Chinese Medicine (TCM) Hospital, Jiaxing, Zhejiang, China
| | - Xiaomin Yang
- Department of Gynecology, Jiaxing University Affiliated Traditional Chinese Medicine (TCM) Hospital, Jiaxing, Zhejiang, China
| | - Yuanxin Fu
- Department of Acupuncture and Massage, Jiaxing University Affiliated Traditional Chinese Medicine (TCM) Hospital, Jiaxing, Zhejiang, China
| | - Yinmin Zhang
- Department of Pediatrics, Jiaxing University Affiliated Traditional Chinese Medicine (TCM) Hospital, Jiaxing, Zhejiang, China
| |
Collapse
|
6
|
Wang X, Xiong X. Mitochondrial Reactive Oxygen Species (mROS) Generation and Cancer: Emerging Nanoparticle Therapeutic Approaches. Int J Nanomedicine 2025; 20:6085-6119. [PMID: 40385494 PMCID: PMC12085131 DOI: 10.2147/ijn.s510972] [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: 12/07/2024] [Accepted: 04/24/2025] [Indexed: 05/20/2025] Open
Abstract
Mitochondrial reactive oxygen species (mROS) are generated as byproducts of mitochondrial oxidative phosphorylation. Changes in mROS levels are involved in tumorigenesis through their effects on cancer genome instability, sustained cancer cell survival, metabolic reprogramming, and tumor metastasis. Recent advances in nanotechnology offer a promising approach for precise regulation of mROS by either enhancing or depleting mROS generation. This review examines the association between dysregulated mROS levels and key cancer hallmarks. We also discuss the potential applications of mROS-targeted nanoparticles that artificially manipulate ROS levels in the mitochondria to achieve precise delivery of antitumor drugs.
Collapse
Affiliation(s)
- Xinyao Wang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
- Queen Mary School of Nanchang University, Nanchang, People’s Republic of China
| | - Xiangyang Xiong
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
7
|
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
|
8
|
Li C, Gong H, Shi P, Liu S, Zhang Q. Different Forms of Regulated Cell Death in Type-2-Diabetes-Mellitus-Related Osteoporosis: A Focus on Mechanisms and Therapeutic Strategies. Int J Mol Sci 2025; 26:4417. [PMID: 40362655 PMCID: PMC12072526 DOI: 10.3390/ijms26094417] [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: 03/11/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder with a high prevalence and challenging treatment options. It significantly affects the function of various organs, including bones, and imposes substantial social and economic costs. Chronic hyperglycemia, insulin resistance, and abnormalities in glucolipid metabolism can lead to cellular damage within the body. Bone dysfunction represents a significant characteristic of diabetic osteoporosis (DOP). Recent studies confirm that cell death is a critical factor contributing to bone damage. Regulated cell death (RCD) is a highly controlled process that involves numerous proteins and specific signaling cascades. RCD processes, including apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and cuproptosis, may be linked to the dysfunction of bone cells in T2DM. In this review, the cell death types of bone cell populations during the pathogenic process of DOP were explored, and the link between cellular RCD processes and the pathogenesis of DOP was further explored. In addition, the research progress on targeting RCD for DOP was summarized in this paper. This may provide a foundation for additional explorations and drug development, as well as new therapeutic concepts for the clinical management of DOP.
Collapse
Affiliation(s)
- Chenchen Li
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; (C.L.); (P.S.); (S.L.); (Q.Z.)
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, Beijing 100191, China
| | - He Gong
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; (C.L.); (P.S.); (S.L.); (Q.Z.)
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, Beijing 100191, China
| | - Peipei Shi
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; (C.L.); (P.S.); (S.L.); (Q.Z.)
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, Beijing 100191, China
| | - Shuyu Liu
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; (C.L.); (P.S.); (S.L.); (Q.Z.)
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, Beijing 100191, China
| | - Qi Zhang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; (C.L.); (P.S.); (S.L.); (Q.Z.)
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, Beijing 100191, China
| |
Collapse
|
9
|
Lu D, Xia B, Feng T, Qi G, Ma Z. The Role of Cancer Organoids in Ferroptosis, Pyroptosis, and Necroptosis: Functions and Clinical Implications. Biomolecules 2025; 15:659. [PMID: 40427552 PMCID: PMC12108908 DOI: 10.3390/biom15050659] [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: 04/04/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
The enduring prevalence of cancer worldwide constitutes a significant public health challenge, thereby emphasizing the imperative for the development of therapeutic models capable of accounting for the heterogeneity inherent in tumors. In this context, cancer organoids have emerged as powerful tools for studying tumor biology, providing valuable insights into the complex interactions within the tumor microenvironment. Concurrently, research is increasingly focused on non-apoptotic forms of regulated cell death (RCD)-including ferroptosis, pyroptosis, and necroptosis-which exert pivotal influences on cancer development and progression. Cancer organoids not only recapitulate the genetic and phenotypic heterogeneity of the original tumors but also enable more precise investigations into the roles of non-apoptotic RCDs within oncology. This review explores the utility of cancer organoids in delineating the molecular mechanisms underlying RCDs and their implications for cancer biology and treatment responses. By synthesizing recent research findings, it highlights the essential role of organoid models in uncovering the intricate details of non-apoptotic RCDs. Furthermore, it emphasizes promising directions for future research that aim to deepen our understanding of these pathways and their therapeutic potential. The integration of organoid models into investigations of ferroptosis, pyroptosis, and necroptosis provides novel insights into oncogenic mechanisms and facilitates the development of targeted therapeutic strategies. By bridging cancer organoids with human pathophysiology, this approach not only provides a transformative framework for dissecting oncogenic pathways but also enables the design of precision therapeutics that selectively target the molecular machinery underlying non-apoptotic RCDs.
Collapse
Affiliation(s)
- Dingci Lu
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
- Department of Clinical Laboratory, The Second Hospital of Jingzhou, Jingzhou 434000, China
| | - Bingqian Xia
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| | - Tianquan Feng
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| | - Gui Qi
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| | - Zhaowu Ma
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| |
Collapse
|
10
|
Zheng P, Hu Z, Shen Y, Gu L, Ouyang Y, Duan Y, Ji G, Dong B, Lin Y, Wen T, Tian Q, Hou Y, Zhou Q, Sun X, Chen X, Wang KL, Luo S, Wu S, Sun Y, Li M, Xiao L, Wu Q, Meng Y, Liu G, Wang Z, Bai X, Duan S, Ding Y, Bi Y, Wang Y, Li G, Liu X, Lu Z, Wu X, Tang Z, Xu D. PSAT1 impairs ferroptosis and reduces immunotherapy efficacy via GPX4 hydroxylation. Nat Chem Biol 2025:10.1038/s41589-025-01887-3. [PMID: 40281343 DOI: 10.1038/s41589-025-01887-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/14/2025] [Indexed: 04/29/2025]
Abstract
Tumor cells adapt to the inflammatory tumor microenvironment (TME) and develop resistance to immunotherapy, with ferroptosis being a major form of tumor cell death. However, the mechanisms by which tumor cells coordinate TME stimuli and their unique metabolic traits to evade ferroptosis and develop resistance to immunotherapy remain unclear. Here we showed that interferon-γ (IFNγ)-activated calcium/calmodulin-dependent protein kinase II phosphorylates phosphoserine aminotransferase 1 (PSAT1) at serine 337 (S337), allowing it to interact with glutathione peroxidase 4 (GPX4) and stabilize the protein, counteracting ferroptosis. PSAT1 elevates GPX4 stability by promoting α-ketoglutarate-dependent PHD3-mediated GPX4 proline 159 (P159) hydroxylation, disrupting its binding to HSC70 and inhibiting autophagy-mediated degradation. In mice, reconstitution of PSAT1 S337A or GPX4 P159A promotes ferroptosis and suppresses triple-negative breast cancer (TNBC) progression. Blocking PSAT1 pS337 with CPP elevates IFNγ-induced ferroptosis and enhances the efficacy of programmed cell death protein 1 (PD-1) antibodies in TNBC. Additionally, PSAT1-mediated GPX4 hydroxylation correlates with poor immunotherapy outcomes in patients with TNBC, highlighting PSAT1's noncanonical role in suppressing ferroptosis and immunotherapy sensitivity.
Collapse
Affiliation(s)
- Peixiang Zheng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zhiqiang Hu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuli Shen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Lina Gu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuan Ouyang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yuran Duan
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Guimei Ji
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Bofei Dong
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yanni Lin
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ting Wen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qi Tian
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yueru Hou
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Sun
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaohan Chen
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | | | - Shudi Luo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Shiqi Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuening Sun
- Department of Pharmacy, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Min Li
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Liwei Xiao
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ying Meng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Guijun Liu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zheng Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shengzhong Duan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanli Bi
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Yuhao Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Gaopeng Li
- Department of Colorectal Surgery and Oncology of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoguang Liu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Xiaohong Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China.
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Daqian Xu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China.
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China.
| |
Collapse
|
11
|
Singh M, Arora HL, Naik R, Joshi S, Sonawane K, Sharma NK, Sinha BK. Ferroptosis in Cancer: Mechanism and Therapeutic Potential. Int J Mol Sci 2025; 26:3852. [PMID: 40332483 PMCID: PMC12028135 DOI: 10.3390/ijms26083852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer drug resistance occurs when cancer cells evade cell death following treatment with chemotherapy, radiation therapy, and targeted therapies. This resistance is often linked to the reprogramming of programmed cell death (PCD) pathways, allowing cancer cells to survive drug-induced stress. However, certain anticancer therapies, when combined with specific agents or inhibitors, can induce ferroptosis-a form of cell death driven by iron-dependent lipid peroxidation. Currently, extensive preclinical and clinical research is underway to investigate the molecular, cellular, and tissue-specific mechanisms underlying ferroptosis, with the goal of identifying strategies to overcome drug resistance in cancers unresponsive to conventional PCD pathways. By harnessing ferroptosis, cancer cells can be compelled to undergo lipid peroxidation-induced death, potentially improving therapeutic outcomes in patients with cancer. This short review aims to enhance the understanding of ferroptosis inducers in cancer therapy and stimulate further research into ferroptosis-based approaches for more effective clinical cancer treatment.
Collapse
Affiliation(s)
- Mansaa Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Hasmiq L. Arora
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Rutuja Naik
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Shravani Joshi
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Kaveri Sonawane
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Birandra K. Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
12
|
Zhang X, Du J, Lin X, Zhang S, Zeng T, Chen M, Huang G, Chen C, Zheng B. Identification of disulfidptosis in esophageal squamous cell carcinoma based on single-cell and bulk RNA-seq data to predict prognosis and treatment response. Front Immunol 2025; 16:1567793. [PMID: 40303412 PMCID: PMC12037556 DOI: 10.3389/fimmu.2025.1567793] [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: 01/28/2025] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Purpose Our study aims to identify the molecular subtypes of genes associated with disulfidptosis in esophageal squamous cell carcinoma(ESCC), develop a prognostic model, and identify potential therapeutic targets. Methods Based on the GSE53625 expression profile data, we identified molecular subtypes with significant survival differences through consensus cluster analysis. Subsequently, univariate Cox, multivariate Cox, and LASSO-Cox regression analysis were used to establish risk stratification models. The transcriptome data of the TCGA-ESCC cohort and the GSE160269 single-cell sequencing dataset were integrated to verify the biological significance of the model, and further analyze the heterogeneity of the tumor immune microenvironment, explore the differences in the intercellular communication network, and screen potential targeted drugs, providing a theoretical basis for subsequent translational research. Results We identified two distinct patterns of disulfidptosis expression with significant differences in overall survival. Then, we constructed the prognostic signature of disulfidptosis, and results showed patients with high score had worse prognosis. Univariate and multivariate Cox analysis demonstrated that the constructed prognostic signature was an independent prognostic factor and was validated in an independent validation set. The two subgroups differed in the proportion of immune cell infiltration and related signaling pathways in ESCC. The exploration of immunotherapy data confirmed our prognostic signature also had certain predictive power for immunotherapy. Drug screening suggested AZD8186 and JQ1 as potential therapies for high-score patients. Conclusion This study provides a new prognostic signature for ESCC, explores new therapeutic targets, and provides new theoretical support for personalized treatment.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Jianting Du
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Xiao Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Shuliang Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Taidui Zeng
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Maohui Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Guanglei Huang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Bin Zheng
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| |
Collapse
|
13
|
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
|
14
|
Zhao S, Yang C, Wan W, Yuan S, Wei H, Chen J. Computational Identification and Validation of Metabolic Cell Death-Related Prognostic Biomarkers for Personalized Treatment Strategies in Prostate Cancer. Cell Biochem Biophys 2025:10.1007/s12013-025-01746-x. [PMID: 40210782 DOI: 10.1007/s12013-025-01746-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 04/12/2025]
Abstract
Prostate cancer (PCa) is a prevalent malignancy characterized by metabolic dysregulation and varied prognosis. Identifying prognostic biomarkers related to metabolic cell death could enhance risk stratification and treatment strategies. The purpose of this study was to identify prognostic genes associated with metabolic cell death in PCa and formulate a risk model for improved patient stratification. We identified genes that exhibit differential expression in The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA-PRAD) cohort (n = 394), with validation using GSE70769 (n = 92) and RT-qPCR on tissue samples from 5 patients. Candidate genes were intersected with metabolic cell death-related genes to identify prognostic markers. Independent prognostic factors were determined utilizing univariate and multivariate Cox regression analyses (p < 0.05, HR ≠ 1). A nomogram was designed, and the validation of gene expression was carried out using RT-qPCR on tissue samples from five PCa patients. A total of 78 candidate genes were identified, with ASNS and ZNF419 emerging as independent prognostic factors. The gene-based risk model successfully stratified patients into high- and low-risk groups, demonstrating correlations with overall survival and clinicopathological features, while also revealing significant differences in immune cell infiltration patterns through immune microenvironment analysis. Additionally, somatic mutation analysis indicated TP53, TTN, and SPOP as frequently mutated genes. This study identifies ASNS and ZNF419 as novel prognostic biomarkers in PCa, contributing to improved risk stratification and personalized treatment strategies. Further investigation into their functional roles may provide insights into therapeutic targets for PCa management.
Collapse
Affiliation(s)
- Shixian Zhao
- Department of urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650033, China
- Kunming Medical University, Kunming, 650500, China
| | - Chadanfeng Yang
- Department of urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650033, China
- Kunming Medical University, Kunming, 650500, China
| | - Weiming Wan
- Department of urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650033, China
| | - Shunhui Yuan
- Department of urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650033, China
| | - Hairong Wei
- Department of urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650033, China.
| | - Jian Chen
- Department of urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650033, China.
| |
Collapse
|
15
|
Lei G, Sun M, Cheng J, Ye R, Lu Z, Horbath A, Huo D, Wu S, Alapati A, Aggarwal S, Xu Z, Mao C, Yan Y, Yao J, Li Q, Chen X, Lee H, Zhuang L, Jiang D, Pataer A, Roth JA, Navin N, Koong AC, You MJ, Lin SH, Gan B. Radiotherapy promotes cuproptosis and synergizes with cuproptosis inducers to overcome tumor radioresistance. Cancer Cell 2025:S1535-6108(25)00132-1. [PMID: 40215978 DOI: 10.1016/j.ccell.2025.03.031] [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: 08/01/2024] [Revised: 01/10/2025] [Accepted: 03/27/2025] [Indexed: 05/15/2025]
Abstract
Cuproptosis is a recently identified form of copper-dependent cell death. Here, we reveal that radiotherapy (RT) induces cuproptosis in cancer cells, independent of apoptosis and ferroptosis, and depletes lipoylated proteins and iron-sulfur (Fe-S) cluster proteins-both hallmarks of cuproptosis-in patient tumors. Mechanistically, RT elevates mitochondrial copper levels by upregulating copper transporter 1 (CTR1) and depleting mitochondrial glutathione, a copper chelator, thereby triggering cuproptosis. Integrated analyses of RNA sequencing (RNA-seq) from radioresistant esophageal cancer cells and single-cell RNA-seq from esophageal tumors of patients unresponsive to RT link radioresistance to the downregulation of BTB and CNC homology 1 (BACH1). This downregulation de-represses the expression of copper-sequestering metallothionein (MT) 1E/X, thereby mitigating cuproptosis and contributing to radioresistance. Copper ionophore treatment sensitizes radioresistant cancer cells and cell line- and patient-derived xenografts to RT by potentiating cuproptosis. Our findings unveil a link between RT and cuproptosis and inform a therapeutic strategy to overcome tumor radioresistance by targeting cuproptosis.
Collapse
Affiliation(s)
- Guang Lei
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mingchuang Sun
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Cheng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rui Ye
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhengze Lu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amber Horbath
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David Huo
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shengrong Wu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anagha Alapati
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sadhna Aggarwal
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhihao Xu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chao Mao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuelong Yan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Yao
- Department of Molecular and Cellular Oncology, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qidong Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiong Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hyemin Lee
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dadi Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Apar Pataer
- Department of Thoracic and Cardiovascular Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas Navin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Albert C Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mingjian James You
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
16
|
Lu W, Zhang Z, Qiao G, Zou G, Li G. Immune Regulation and Disulfidptosis in Atherosclerosis Influence Disease Progression and Therapy. Biomedicines 2025; 13:926. [PMID: 40299531 PMCID: PMC12025079 DOI: 10.3390/biomedicines13040926] [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: 02/11/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Atherosclerosis is a progressive and complex vascular pathology characterized by cellular heterogeneity, metabolic dysregulation, and chronic inflammation. Despite extensive research, the intricate molecular mechanisms underlying its development and progression remain incompletely understood. Methods: Single-cell RNA sequencing (scRNA-seq) was employed to conduct a comprehensive mapping of immune cell enrichment and interactions within atherosclerotic plaques, aiming to investigate the cellular and molecular complexities of these structures. This approach facilitated a deeper understanding of the heterogeneities present in smooth muscle cells, which were subsequently analyzed using pseudotime trajectory analysis to monitor the developmental trajectories of smooth muscle cell (SMC) subpopulations. An integrative bioinformatics approach, primarily utilizing Weighted Gene Co-expression Network Analysis (WGCNA) and machine learning techniques, identified Cathepsin C (CTSC), transforming growth factor beta-induced protein (TGFBI), and glia maturation factor-γ (GMFG) as critical biomarkers. A diagnostic risk score model was developed and rigorously tested through Receiver Operating Characteristic analysis. To illustrate the functional impact of CTSC on the regulation of plaque formation and SMC viability, both in vitro and in vivo experimental investigations were conducted. Results: An analysis revealed SMCs identified as the most prominent cellular type, exhibiting the highest density of disulfidptosis. Pseudotime trajectory analysis illuminated the dynamic activation pathways in SMCs, highlighting their significant role in plaque development and instability. Further characterization of macrophage subtypes demonstrated intercellular communication with SMCs, which exhibited specific signaling pathways, particularly between the proximal and core areas of plaques. The integrated diagnostic risk score model, which incorporates CTSC, TGFBI, and GMFG, proved to be highly accurate in distinguishing high-risk patients with elevated immune responses and systemic inflammation. Knockdown experiments of CTSC conducted in vitro revealed enhanced SMC survival rates, reduced oxidative stress, and inhibited apoptosis, while in vivo experiments confirmed a decrease in plaque burden and improvement in lipid profiles. Conclusions: This study emphasizes the significance of disulfidptosis in the development of atherosclerosis and identifies CTSC as a potential therapeutic target for stabilizing plaques by inhibiting SMC apoptosis and oxidative damage. Additionally, the risk score model serves as a valuable diagnostic tool for identifying high-risk patients and guiding precision treatment strategies.
Collapse
Affiliation(s)
| | - Zhidong Zhang
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 451460, China; (W.L.); (G.Q.); (G.Z.); (G.L.)
| | | | | | | |
Collapse
|
17
|
Rajabi F, Hajipour-Verdom B, Abdolmaleki P. Static magnetic field promotes the doxorubicin toxicity effects on osteosarcoma cells. Sci Rep 2025; 15:11902. [PMID: 40195518 PMCID: PMC11977194 DOI: 10.1038/s41598-025-96802-0] [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: 08/17/2024] [Accepted: 03/31/2025] [Indexed: 04/09/2025] Open
Abstract
Osteosarcoma, a highly aggressive bone cancer, primarily affects adolescents and is frequently treated with conventional chemotherapy, such as doxorubicin (DOX). However, the efficacy of DOX is often limited by severe side effects and drug resistance. This study investigates the synergistic effects of static magnetic fields (SMF) and DOX on G292 osteosarcoma cells and HFF normal fibroblasts. Cell viability was assessed using the MTT assay, intracellular reactive oxygen species (ROS) levels were quantified via DCFDA staining and flow cytometry, and iron and calcium homeostasis were analyzed using ICP-OES. Apoptosis and necrosis were determined through Annexin V-FITC/PI staining. Results demonstrated that the combination of SMF and DOX significantly reduced G292 cell viability compared to DOX alone, with IC50 values decreased from 3.2 µM (at 3 mT, p < 0.01) to 0.8 µM (at 24 mT, p < 0.001) at 24 h. Apoptosis rates increased from 8.12% with DOX alone to 16% with SMF + DOX. While DOX alone elevated ROS levels by 59.15% in G292 cells, SMF further amplified apoptosis by enhancing ROS generation and disrupting iron and calcium homeostasis. These findings suggest that SMF enhances DOX-induced cytotoxicity in osteosarcoma cells by promoting ROS production, altering metal ion homeostasis, and increasing apoptosis. SMF represents a promising adjuvant therapy for osteosarcoma treatment, though further in vivo studies are necessary to optimize treatment parameters and evaluate clinical applicability.
Collapse
Affiliation(s)
- Fatemeh Rajabi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154, Tehran, Iran
| | - Behnam Hajipour-Verdom
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154, Tehran, Iran
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), 1517964311, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154, Tehran, Iran.
| |
Collapse
|
18
|
Ning L, Li X, Xu Y, Si Y, Zhao H, Ren Q. Multi-Omics Analysis Revealed That TAOK1 Can Be Used as a Prognostic Marker and Target in a Variety of Tumors, Especially in Cervical Cancer. Onco Targets Ther 2025; 18:335-353. [PMID: 40109409 PMCID: PMC11920640 DOI: 10.2147/ott.s506582] [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: 11/14/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Background Thousand and One Kinase 1 (TAOK1), a member of the MAPK kinase family, plays a crucial role in processes like microtubule dynamics, DNA damage response, and neurodevelopment. While TAOK1 is linked to tumorigenesis, its oncogenic role across cancers remains unclear. This study aims to explore the relationship between TAOK1 expression, prognosis, and immune function in various cancers. Methods We analyzed TAOK1 expression in multiple cancers using TCGA, GEO, CCLE, and other bioinformatics databases. The correlation between TAOK1 expression and immune cell infiltration was assessed with the ESTIMATE algorithm. We also examined associations with tumor stemness, DNA methylation, gene copy number alterations, and drug sensitivity. The oncogenic role of TAOK1 was further evaluated in vitro with SiHa and A2780 cells and in vivo with TAOK1 overexpression in SiHa cells. Results TAOK1 is a key prognostic biomarker in various cancers and its high expression is associated with poor prognosis. It showed a significant negative correlation with immune cell infiltration and immune checkpoints. GSEA identified its involvement in key tumour pathways, highlighting the therapeutic potential of inhibiting the TAOK1 gene. The high expression of TAOK1 is associated with DNA methylation and gene copy number variation, and in addition its upstream regulator, EP300, is closely associated with TAOK1 expression. In vitro cellular experiments demonstrated that inhibition of TAOK1 reduced the proliferation of SiHa and A2780 cells, whereas overexpression of TAOK1 in SiHa cells promoted growth. These findings were further validated in vivo by nude mouse tumourigenicity assay and human tissue immunohistochemistry. Conclusion TAOK1 serves as a promising prognostic biomarker and potential therapeutic target, especially for cervical cancer. These results support its clinical potential in cancer prognosis and treatment strategies.
Collapse
Affiliation(s)
- Li Ning
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Xiu Li
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Yating Xu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Yu Si
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Hongting Zhao
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Qingling Ren
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
19
|
Seo R, de Guzman ACV, Park S, Lee JY, Kang SJ. Cancer-intrinsic Cxcl5 orchestrates a global metabolic reprogramming for resistance to oxidative cell death in 3D. Cell Death Differ 2025:10.1038/s41418-025-01466-y. [PMID: 40050422 DOI: 10.1038/s41418-025-01466-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 01/10/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Pancreatic ductal adenocarcinoma is characterized by a three-dimensional (3D) tumor microenvironment devoid of oxygen and nutrients but enriched in extracellular matrix, which acts as a physical and chemical barrier. In 3D, cancer cells reprogram their metabolic pathways in ways that help them survive hostile conditions. However, little is known about the metabolic phenotypes of cancer cells in 3D and the intrinsic cues that modulate them. We found that Cxcl5 deletion restricted pancreatic tumor growth in a 3D spheroid-in-Matrigel culture system without affecting cancer cell growth in 2D culture. Cxcl5 deletion impaired 3D-specific global metabolic reprogramming, resistance to hypoxia-induced cell death, and upregulation of Hif1α and Myc. Overexpression of Hif1α and Myc, however, effectively restored 3D culture-induced metabolic reconfiguration, growth, redox homeostasis, and mitochondrial function in Cxcl5-/- cells, reducing ferroptosis. We also found that pancreatic cancer patients with higher expression of hypoxia and metabolism-related genes whose expression is well-correlated with CXCL5 generally have poorer prognosis. Together, our findings identify an unanticipated role of Cxcl5 in orchestrating the cancer metabolic reprogramming in 3D culture that is required for energy and biomass maintenance and that restricts oxidative cell death. Thus, our results provide a rationale for targeting CXCL5 as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Ramin Seo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Arvie Camille V de Guzman
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sunghyouk Park
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji Youn Lee
- Biometrology Group, Division of Biomedical Metrology, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Suk-Jo Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
20
|
Fu M, Wu H, Peng P, Wang J, Cao D. Metabolism-Related Programmed Cell Death: Unveiling Prognostic Biomarkers, Immune Checkpoints, and Therapeutic Strategies in Ovarian Cancer. Cancer Invest 2025; 43:183-204. [PMID: 40192346 DOI: 10.1080/07357907.2025.2481436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/18/2025] [Accepted: 03/15/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Ovarian cancer (OC), the gynecologic malignancy with the poorest prognosis, is driven by metabolic reprogramming and dysregulated programmed cell death (PCD). However, their interplay and prognostic significance remain inadequately understood. METHODS Transcriptomic data from OC patients and healthy controls (TCGA and GTEx) were analyzed to identify differentially expressed genes (DEGs) intersecting with metabolism-related (MRGs) and PCD-related genes (PCDRGs). Prognostic genes were determined using univariate Cox regression, LASSO, multivariate Cox regression, and stepwise analyses. Consensus clustering revealed enrichment differences, while a risk model and nomogram were developed for outcome prediction. Associations between prognostic genes, immune microenvironment, and drug sensitivity were also assessed. RESULTS A total of 166 candidate genes were identified, with PLA2G2D, LPCAT3, ARG1, PLA2G4A, and EXOSC3 emerging as significant prognostic markers. The risk model demonstrated marked survival differences, while the nomogram showed robust calibration for survival prediction. Differential immune cell infiltration was observed between risk groups. Additionally, Sinularin and Fulvestrant exhibited variable sensitivity, validated through molecular docking models. CONCLUSION Metabolism-related PCD genes were identified as pivotal prognostic markers in OC, providing critical insights for prognostic evaluation and targeted therapy development.
Collapse
Affiliation(s)
- Mengdi Fu
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hao Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Peng
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinhui Wang
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Dongyan Cao
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
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
|
22
|
Luo JY, Deng YL, Lu SY, Chen SY, He RQ, Qin DY, Chi BT, Chen G, Yang X, Peng W. Current Status and Future Directions of Ferroptosis Research in Breast Cancer: Bibliometric Analysis. Interact J Med Res 2025; 14:e66286. [PMID: 40009842 PMCID: PMC11904379 DOI: 10.2196/66286] [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: 09/09/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Ferroptosis, as a novel modality of cell death, holds significant potential in elucidating the pathogenesis and advancing therapeutic strategies for breast cancer. OBJECTIVE This study aims to comprehensively analyze current ferroptosis research and future trends, guiding breast cancer research advancements and innovative treatment strategies. METHODS This research used the R package Bibliometrix (Department of Economic and Statistical Sciences at the University of Naples Federico II), VOSviewer (Centre for Science and Technology Studies at Leiden University), and CiteSpace (Drexel University's College of Information Science and Technology), to conduct a bibliometric analysis of 387 papers on breast cancer and ferroptosis from the Web of Science Core Collection. The analysis covers authors, institutions, journals, countries or regions, publication volumes, citations, and keywords. RESULTS The number of publications related to this field has surged annually, with China and the United States collaborating closely and leading in output. Sun Yat-sen University stands out among the institutions, while the journal Frontiers in Oncology and the author Efferth T contribute significantly to the field. Highly cited papers within the domain primarily focus on the induction of ferroptosis, protein regulation, and comparisons with other modes of cell death, providing a foundation for breast cancer treatment. Keyword analysis highlights the maturity of glutathione peroxidase 4-related research, with breast cancer subtypes emerging as motor themes and the tumor microenvironment, immunotherapy, and prognostic models identified as basic themes. Furthermore, the application of nanoparticles serves as an additional complement to the basic themes. CONCLUSIONS The current research status in the field of ferroptosis and breast cancer primarily focuses on the exploration of relevant theoretical mechanisms, whereas future trends and mechanisms emphasize the investigation of therapeutic strategies, particularly the clinical application of immunotherapy related to the tumor microenvironment. Nanotherapy has demonstrated significant clinical potential in this domain. Future research directions should deepen the exploration in this field and accelerate the clinical translation of research findings to provide new insights and directions for the innovation and development of breast cancer treatment strategies.
Collapse
Affiliation(s)
- Jia-Yuan Luo
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu-Long Deng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shang-Yi Lu
- Department of Hepatological and Gland Surgery, Wuzhou Gongren Hospital/The Seventh Affiliated Hospital of Guangxi Medical University, Wuzhou, China
| | - Si-Yan Chen
- Day Chemotherapy Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rong-Quan He
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Di-Yuan Qin
- Department of Computer Science and Technology, School of Computer and Electronic Information, Guangxi University, Nanning, China
| | - Bang-Teng Chi
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xia Yang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wei Peng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
23
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [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
|
24
|
Chen Y, Zhang D, Yang H, Wu J, He W. Advances in the study of disulfidptosis in digestive tract tumors. Discov Oncol 2025; 16:186. [PMID: 39954025 PMCID: PMC11829889 DOI: 10.1007/s12672-025-01875-y] [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: 11/19/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
Disulfidptosis, a recently identified cell death mechanism, plays a pivotal role in the development, progression, and treatment of digestive tract tumors, including gastric cancer, hepatocellular cancer, esophageal cancer, colorectal cancer, pancreatic cancer, cholangiocarcinoma, and neuroendocrine tumors, which have high global incidence and mortality rates. Analyzing the expression of disulfidptosis-related gene expression within the tumor microenvironment enhances our understanding of tumor biology and facilitates novel diagnostic and therapeutic strategies. Research on immune infiltration and checkpoints can identify therapeutic targets linked to disulfidptosis, thereby improving immunotherapy efficacy. Targeting genes such as SLC7A11, which are essential for maintaining glutathione levels and regulating oxidative stress, may overcome chemoresistance and enhance existing treatments. Disulfidptosis could complement current therapies as it induces cytoskeletal collapse and selective tumor cell death, especially in chemoresistant cancers. Additionally, genes like SLC7A11, RPN1, and NCKAP1 in gastric cancer correlate with poor prognosis, highlighting their potential as prognostic biomarkers. Personalized medicine approaches utilizing disulfidptosis-related biomarkers could identify patients who would benefit from therapies targeting oxidative stress regulation, leading to more precise treatments and improved outcomes. This review summarizes disulfidptosis mechanisms, advancements in digestive cancers, and the potential of related genes for prognosis, immune response evaluation, and targeted therapies, providing novel perspectives for diagnosis and personalized treatment.
Collapse
Affiliation(s)
- Yue Chen
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, Jiangsu Province, China
| | - Dachuan Zhang
- Department of Pathology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, Jiangsu Province, China
| | - Haonan Yang
- Department of Hematology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jun Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, Jiangsu Province, China
| | - Wenting He
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, Jiangsu Province, China.
| |
Collapse
|
25
|
Zhang X, Wang R, Zhang X, Yang Y, Tian R. Ferroptosis related CPT1A and GDF15 gene polymorphisms are risk factors for lung adenocarcinoma: A case-control study. Gene 2025; 933:149002. [PMID: 39401734 DOI: 10.1016/j.gene.2024.149002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/28/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Ferroptosis is not only a consequence of inflammation, but also a dynamic process. Recent bioinformatics analysis suggests that ferroptosis related genes might be associated with lung adenocarcinoma (LUAD). CPT1A and GDF15 are critical for the process of ferroptosis and development of inflammation; however, little study focused on the mutation level of these genes in patients with LUAD. METHODS The candidate SNPs in CPT1A and GDF15 were genotyped in 320 pairs of LUAD patients and controls using Mass ARRAY platform. Moreover, the different expression of CPT1A and GDF15 in LUAD cases and healthy controls were validated by qRT-PCR and ELISA. RESULTS The rs80356779 G > A, rs3019594 C > T, rs888663 T > G and rs4808793 G > C all exhibited an increased risk of the disease (p < 0.05). Moreover, the rs80356779-GA, rs3019594-TT, rs888663-TG and rs4808793-CC genotypes were all related to different levels of increase in LUAD risk (p < 0.05). Genetic model results showed that rs80356779 G > A, rs888663 T > G and rs4808793 G > C were associated with LUAD susceptibility under dominant and additive models (p < 0.05), while rs3019594 C > T was correlated with an elevated risk of the disease in all three models (p < 0.05). Additionally, patients with rs80356779 G > A and rs3019594 C > T exhibited lower expression and serum concentration of CPT1A compared with wile types, and patients with rs888663 T > G and rs4808793 G > C exhibited higher serum and expression level of GDF15. CONCLUSION The results provided new clues for the role of ferroptosis in LUAD and new potential targets for screening of susceptible population.
Collapse
Affiliation(s)
- Xing Zhang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Rong Wang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xia Zhang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Yanli Yang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Ruifen Tian
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
26
|
Imam M, Ji J, Zhang Z, Yan S. Targeting the initiator to activate both ferroptosis and cuproptosis for breast cancer treatment: progress and possibility for clinical application. Front Pharmacol 2025; 15:1493188. [PMID: 39867656 PMCID: PMC11757020 DOI: 10.3389/fphar.2024.1493188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/12/2024] [Indexed: 01/28/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide. Metal metabolism is pivotal for regulating cell fate and drug sensitivity in breast cancer. Iron and copper are essential metal ions critical for maintaining cellular function. The accumulation of iron and copper ions triggers distinct cell death pathways, known as ferroptosis and cuproptosis, respectively. Ferroptosis is characterized by iron-dependent lipid peroxidation, while cuproptosis involves copper-induced oxidative stress. They are increasingly recognized as promising targets for the development of anticancer drugs. Recently, compelling evidence demonstrated that the interplay between ferroptosis and cuproptosis plays a crucial role in regulating breast cancer progression. This review elucidates the converging pathways of ferroptosis and cuproptosis in breast cancer. Moreover, we examined the value of genes associated with ferroptosis and cuproptosis in the clinical diagnosis and treatment of breast cancer, mainly outlining the potential for a co-targeting approach. Lastly, we delve into the current challenges and limitations of this strategy. In general, this review offers an overview of the interaction between ferroptosis and cuproptosis in breast cancer, offering valuable perspectives for further research and clinical treatment.
Collapse
Affiliation(s)
| | | | | | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
27
|
Gu Q, Wang Y, Yi P, Cheng C. Theoretical framework and emerging challenges of lipid metabolism in cancer. Semin Cancer Biol 2025; 108:48-70. [PMID: 39674303 DOI: 10.1016/j.semcancer.2024.12.002] [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: 10/11/2024] [Revised: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Elevated lipid metabolism is one of hallmarks of malignant tumors. Lipids not only serve as essential structural components of biological membranes but also provide energy and substrates for the proliferation of cancer cells and tumor growth. Cancer cells meet their lipid needs by coordinating the processes of lipid absorption, synthesis, transport, storage, and catabolism. As research in this area continues to deepen, numerous new discoveries have emerged, making it crucial for scientists to stay informed about the developments of cancer lipid metabolism. In this review, we first discuss relevant concepts and theories or assumptions that help us understand the lipid metabolism and -based cancer therapies. We then systematically summarize the latest advancements in lipid metabolism including new mechanisms, novel targets, and up-to-date pre-clinical and clinical investigations of anti-cancer treatment with lipid metabolism targeted drugs. Finally, we emphasize emerging research directions and therapeutic strategies, and discuss future prospective and emerging challenges. This review aims to provide the latest insights and guidance for research in the field of cancer lipid metabolism.
Collapse
Affiliation(s)
- Qiuying Gu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Chunming Cheng
- Department of Oncology Science, OU Health Stephenson Cancer Center at University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
28
|
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
|
29
|
Gong K, Zheng Y, Liu Y, Zhang T, Song Y, Chen W, Guo L, Zhou J, Liu W, Fang T, Chen Y, Wang J, Pan F, Shi K. Phosphocholine inhibits proliferation and reduces stemness of endometrial cancer cells by downregulating mTOR-c-Myc signaling. Cell Mol Life Sci 2024; 82:3. [PMID: 39680126 PMCID: PMC11649893 DOI: 10.1007/s00018-024-05517-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Endometrial cancer (EC) represents a serious health concern among women globally. Excessive activation of the protooncogene c-Myc (c-Myc) is associated with the proliferation and stemness of EC cells. Phosphocholine (PC), which is synthesized by choline kinase alpha (CHKA) catalysis, is upregulated in EC tumor tissues. The present study aimed to investigate the effect of PC accumulation on EC cells and clarify the relationship between PC accumulation and c-Myc activity in EC. METHODS The c-Myc and CHKA expression in EC tumor tissues were examined using immunohistochemistry. Cell Counting Kit-8 assay, colony formation assay, flow cytometry, western blotting, BrdU staining, and tumorsphere formation assay were used to assess the effect of PC accumulation on EC cells. The mechanism by which PC accumulation inhibits c-Myc was evaluated using RNA-sequencing. Patient-derived organoid (PDO) models were utilised to explore the preclinical efficacy of PC against EC cells. RESULTS PC accumulation suppressed EC cell proliferation and stemness by inhibiting the activation of the mammalian target of rapamycin (mTOR)-c-Myc signaling. PC accumulation promoted excessive reactive oxygen species production, which reduced the expression of GTPase HRAS. This, in turn, inhibited the mTOR-c-Myc axis and induced EC cell apoptosis. Finally, PC impeded proliferation and downregulated the expression of the mTOR-MYC signaling in EC PDO models. CONCLUSIONS PC accumulation impairs the proliferation ability and stem cell characteristics of EC cells by inhibiting the activated mTOR-c-Myc axis, potentially offering a promising strategy to enhance the efficacy of EC clinical therapy through the promotion of PC accumulation in tumor cells.
Collapse
Affiliation(s)
- Kunxiang Gong
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yanqin Zheng
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yaqiong Liu
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Tiansong Zhang
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Yiming Song
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Weiwei Chen
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Lirong Guo
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jie Zhou
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Wenjie Liu
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Tianlin Fang
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Yun Chen
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jingyao Wang
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Feifei Pan
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Kun Shi
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
30
|
Hu J, Zhu J, Chen T, Zhao Y, Xu Q, Wang Y. Cuproptosis in cancer therapy: mechanisms, therapeutic application and future prospects. J Mater Chem B 2024; 12:12191-12206. [PMID: 39526989 DOI: 10.1039/d4tb01877j] [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: 11/16/2024]
Abstract
Cuproptosis is a regulated form of cell death induced by the accumulation of metal ions and is closely linked to aspects of cellular drug resistance, cellular metabolism, and signalling pathways. Due to its crucial role in regulating physiological and pathological processes, cuproptosis has gained increasing significance as a potential target for anticancer drug development. In this review, we introduce the definition of cuproptosis and provide a comprehensive discussion of the mechanisms of cuproptosis. In addition, the methods for the detection of cuproptosis are summarized, and recent advances in cuproptosis in cancer therapy are reviewed, mainly in terms of elesclomol (ES)-mediated cuproptosis and disulfiram (DSF)-mediated cuproptosis, which provided practical value for applications. Finally, the current challenges and future development of cuproptosis-mediated cancer therapy are discussed. In summary, this review highlights recent progress on cuproptosis in cancer therapy, offering novel ideas and strategies for future research and applications.
Collapse
Affiliation(s)
- Jiawei Hu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Junfei Zhu
- China-Japan Friendship Hospital, No. 2 Sakura East Street, Chaoyang District, Beijing, China
| | - Tao Chen
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yudie Zhao
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Qingwen Xu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| |
Collapse
|
31
|
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
|
32
|
Abida, Altamimi ASA, Ghaboura N, Balaraman AK, Rajput P, Bansal P, Rawat S, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H, Deb PK. Therapeutic Potential of lncRNAs in Regulating Disulfidptosis for Cancer Treatment. Pathol Res Pract 2024; 263:155657. [PMID: 39437641 DOI: 10.1016/j.prp.2024.155657] [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: 09/02/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
Non-coding RNAs (lncRNAs) play critical roles in various cellular processes, including a novel form of regulated cell death known as disulfidptosis, characterized by accumulating protein disulfide bonds and severe endoplasmic reticulum stress. This review highlights the therapeutic potential of lncRNAs in regulating disulfidptosis for cancer treatment, emphasizing their influence on key pathway components such as GPX4, SLC7A11, and PDIA family members. Recent studies have demonstrated that targeting specific lncRNAs can sensitize cancer cells to disulfidptosis, offering a promising approach to cancer therapy. The regulation of disulfidptosis by lncRNAs involves various signaling pathways, including oxidative stress, ER stress, and calcium signaling. This review also discusses the molecular mechanisms underlying lncRNA regulation of disulfidptosis, the challenges of developing lncRNA-based therapies, and the future potential of this rapidly advancing field in cancer research.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmacy Practice, Pharmacy Program, Batterjee Medical College, PO Box 6231, Jeddah 21442, Saudi Arabia
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Pranchal Rajput
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India.
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Fadiyah Jadid Alanazi
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Al Jouf 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Center for Global health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Pran Kishore Deb
- Department of Pharmaceutical Sciences and Technology, Birla Institue of Technology (BIT), Mesra, Ranchi, Jharkhand 835215, India
| |
Collapse
|
33
|
Yang J, Gao Y, Mao H, Kuang X, Tian F. Qiju Dihuang Pill protects the lens epithelial cells via alleviating cuproptosis in diabetic cataract. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118444. [PMID: 38851473 DOI: 10.1016/j.jep.2024.118444] [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: 04/21/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qiju Dihuang Pill (QDP) is a traditional Chinese medicine prescription for the treatment of eye diseases. Novel literature reports that copper-induced cell death, called as cuproptosis, is a copper-dependent and differs distinctly from other types of cell death. AIM OF THE STUDY The present study aims to investigate whether QDP could protect lens epithelial cells via alleviating copper-induced death in diabetic cataract. MATERIALS AND METHODS The different concentration of QDP medicated serum was administrated on high glucose (HG)-induced human lens epithelial cells (HLECs). The copper concentration was tested using Elabscience Copper Assay kit. The proliferation was detected using CCK-8 and EdU assays. The molecular binding was identified using RIP-PCR and luciferase reporter assay. RESULTS Results indicated that HG culture condition triggered the copper concentration and repressed the proliferation of HLECs. Then, the elesclomol-Cu (Es-Cu) administration up-regulated the copper concentration and inhibited the proliferation, and cuproptosis inhibitor tetrathiomolybdate (TTM) could specifically reverse the consequence. QDP treatment reduced the copper concentration and cuproptosis-related genes (SLC31A1, FDX1). MeRIP-Seq and RIP-PCR confirmed that QDP reduced the stability of SLC31A1 mRNA through m6A modified site, and copper actually synergized the molecular binding efficiency. Rescue assay verified the role of QDP and SLC31A1 on HLECs' cuproptosis characteristic. CONCLUSION This research identified the protective role of QDP on HG-induced HLECs in DC through decreasing m6A/SLC31A1-mediated cuproptosis in DC. This finding provides novel insights into mechanisms for QDP and sheds light on the multifaceted role of traditional prescription on DC.
Collapse
Affiliation(s)
- Jun Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Yichen Gao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Han Mao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Xinqi Kuang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Fang Tian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China.
| |
Collapse
|
34
|
Aquilano K, Filomeni G, Faraonio R, De Luca A. Editorial: Metals in cancer: from intracellular signaling to therapy. Front Oncol 2024; 14:1495825. [PMID: 39450254 PMCID: PMC11499201 DOI: 10.3389/fonc.2024.1495825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Affiliation(s)
- Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Redox Biology Research Group, Danish Cancer Institute, Copenhagen, Denmark
| | - Raffaella Faraonio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
35
|
He C, Li Q, Wu W, Liu K, Li X, Zheng H, Lai Y. Ferroptosis-associated genes and compounds in renal cell carcinoma. Front Immunol 2024; 15:1473203. [PMID: 39399506 PMCID: PMC11466770 DOI: 10.3389/fimmu.2024.1473203] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
As the main type of renal cell carcinoma (RCC), clear cell RCC (ccRCC) is often associated with the deletion or mutation of the von Hippel Lindau (VHL) gene, enhancement of glucose and lipid metabolism, and heterogeneity of the tumor microenvironment. VHL alterations in RCC cells lead to the activation of hypoxia-inducible factors and their downstream target vascular endothelial growth factor, and to the reprogramming of multiple cell death pathways and metabolic weakness, including ferroptosis, which are associated with targeted therapy or immunotherapy. The changes in biological metabolites (e.g., iron and lipids) support ferroptosis as a potential therapeutic strategy for RCC, while iron metabolism and ferroptosis regulation have been examined as anti-RCC agents in numerous studies, and various ferroptosis-related molecules have been shown to be related to the metastasis and prognosis of ccRCC. For example, glutathione peroxidase 4 and glutaminase inhibitors can inhibit pyrimidine synthesis and increase reactive oxygen species levels in VHL-deficient RCC cells. In addition, the release of damage-associated molecular patterns by tumor cells undergoing ferroptosis also mediates antitumor immunity, and immune therapy can synergize with targeted therapy or radiotherapy through ferroptosis. However, Inducing ferroptosis not only suppresses cancer, but also promotes cancer development due to its potential negative effects on anti-cancer immunity. Therefore, ferroptosis and various tumor microenviroment-related molecules may co-occur during the development and treatment of RCC, and further understanding of the interactions, core targets, and related drugs of ferroptosis may provide new combination drug strategies for RCC treatment. Here we summarize the key genes and compounds on ferroptosis and RCC in order to envision future treatment strategies and to provide sufficient information for overcoming RCC resistance through ferroptosis.
Collapse
Affiliation(s)
- Chengwu He
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Qingyi Li
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Weijia Wu
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ke Liu
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xingwen Li
- Tibet Future Biomedicine Company Limited, Golmud, Qinghai, China
| | - Hanxiong Zheng
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yongchang Lai
- Department of Pharmaceutical Management, School of Medical Business, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
36
|
Jiang A, Liu W, Liu Y, Hu J, Zhu B, Fang Y, Zhao X, Qu L, Lu J, Liu B, Qi L, Cai C, Luo P, Wang L. DCS, a novel classifier system based on disulfidptosis reveals tumor microenvironment heterogeneity and guides frontline therapy for clear cell renal carcinoma. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:263-279. [PMID: 39281723 PMCID: PMC11401502 DOI: 10.1016/j.jncc.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 04/26/2024] [Accepted: 06/13/2024] [Indexed: 09/18/2024] Open
Abstract
Background Emerging evidence suggests that cell deaths are involved in tumorigenesis and progression, which may be treated as a novel direction of cancers. Recently, a novel type of programmed cell death, disulfidptosis, was discovered. However, the detailed biological and clinical impact of disulfidptosis and related regulators remains largely unknown. Methods In this work, we first enrolled pancancer datasets and performed multi-omics analysis, including gene expression, DNA methylation, copy number variation and single nucleic variation profiles. Then we deciphered the biological implication of disulfidptosis in clear cell renal cell carcinoma (ccRCC) by machine learning. Finally, a novel agent targeting at disulfidptosis in ccRCC was identified and verified. Results We found that disulfidptosis regulators were dysregulated among cancers, which could be explained by aberrant DNA methylation and genomic mutation events. Disulfidptosis scores were depressed among cancers and negatively correlated with epithelial mesenchymal transition. Disulfidptosis regulators could satisfactorily stratify risk subgroups in ccRCC, and a novel subtype, DCS3, owning with disulfidptosis depression, insensitivity to immune therapy and aberrant genome instability were identified and verified. Moreover, treating DCS3 with NU1025 could significantly inhibit ccRCC malignancy. Conclusion This work provided a better understanding of disulfidptosis in cancers and new insights into individual management based on disulfidptosis.
Collapse
Affiliation(s)
- Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Junyi Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baohua Zhu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yu Fang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xuenan Zhao
- Center for Translational Medicine, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Juan Lu
- Vocational Education Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Bing Liu
- Department of Urology, The Third Affiliated Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Chen Cai
- Department of Special Clinic, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
37
|
Qin X, Chen Z, Wu L, Ding R. A disulfidptosis-associated long noncoding RNA signature to predict low-grade glioma classification, prognosis, tumor microenvironment, and therapy regimens: Observational study. Medicine (Baltimore) 2024; 103:e39316. [PMID: 39183405 PMCID: PMC11346906 DOI: 10.1097/md.0000000000039316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/23/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
This study aimed to investigate the function of disulfidptosis-associated long noncoding RNAs (DAlncRNAs) in low-grade gliomas (LGG) through bioinformatics analysis and construct a signature to predict the classification, prognosis, tumor microenvironment, and selection of immunotherapy and chemotherapy in LGG. Genomic, clinical, and mutational information of 526 patients with LGG was retrieved from The Cancer Genome Atlas repository. A nonnegative matrix factorization algorithm was applied to classify patients with LGG. Univariate, LASSO regression, and multivariate Cox regression analyses were performed to determine prognostic DAlncRNAs. Following the median risk score, we defined the sample as a high-risk (HR) or low-risk group. Finally, survival, receiver operating characteristic curve, risk curve, principal component, independent prognosis, risk difference, functional enrichment, tumor microenvironment, immune cell infiltration, mutation, and drug sensitivity analyses were performed. Patients were classified into C1 and C2 subtypes associated with disulfidptosis. Eight prognostic DAlncRNAs (AC003035.2, AC010157.2, AC010273.3, AC011444.3, AC092667.1, AL450270.1, AL645608.2, and LINC01571) were identified, and a prognostic signature of LGG was developed. The DAlncRNA-based signature was found to be an independent prognostic factor in patients with LGG, thereby constructing a nomogram. In addition, in the HR group, immune function was more active and the tumor mutation burden was higher. The patients were mainly composed of subtype C2, and their prognosis was worse. Immunotherapy and chemotherapy were predicted in the HR and low-risk groups, respectively. Our study, based on DAlncRNAs, highlights 2 disulfidptosis-associated LGG subtypes with different prognostic and immune characteristics and creates a novel disulfidptosis-associated prognostic signature, which may inform the classification, prognosis, molecular pathogenesis, and therapeutic strategies for patients with LGG.
Collapse
Affiliation(s)
- Xiaohong Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rui Ding
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
38
|
Wu H, Fu M, Wu M, Cao Z, Zhang Q, Liu Z. Emerging mechanisms and promising approaches in pancreatic cancer metabolism. Cell Death Dis 2024; 15:553. [PMID: 39090116 PMCID: PMC11294586 DOI: 10.1038/s41419-024-06930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Pancreatic cancer is an aggressive cancer with a poor prognosis. Metabolic abnormalities are one of the hallmarks of pancreatic cancer, and pancreatic cancer cells can adapt to biosynthesis, energy intake, and redox needs through metabolic reprogramming to tolerate nutrient deficiency and hypoxic microenvironments. Pancreatic cancer cells can use glucose, amino acids, and lipids as energy to maintain malignant growth. Moreover, they also metabolically interact with cells in the tumour microenvironment to change cell fate, promote tumour progression, and even affect immune responses. Importantly, metabolic changes at the body level deserve more attention. Basic research and clinical trials based on targeted metabolic therapy or in combination with other treatments are in full swing. A more comprehensive and in-depth understanding of the metabolic regulation of pancreatic cancer cells will not only enrich the understanding of the mechanisms of disease progression but also provide inspiration for new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hao Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengdi Fu
- Department of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhen Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qiyao Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
39
|
Zhao R, Sukocheva O, Tse E, Neganova M, Aleksandrova Y, Zheng Y, Gu H, Zhao D, Madhunapantula SV, Zhu X, Liu J, Fan R. Cuproptosis, the novel type of oxidation-induced cell death in thoracic cancers: can it enhance the success of immunotherapy? Cell Commun Signal 2024; 22:379. [PMID: 39068453 PMCID: PMC11282696 DOI: 10.1186/s12964-024-01743-2] [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: 02/26/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Copper is an important metal micronutrient, required for the balanced growth and normal physiological functions of human organism. Copper-related toxicity and dysbalanced metabolism were associated with the disruption of intracellular respiration and the development of various diseases, including cancer. Notably, copper-induced cell death was defined as cuproptosis which was also observed in malignant cells, representing an attractive anti-cancer instrument. Excess of intracellular copper leads to the aggregation of lipoylation proteins and toxic stress, ultimately resulting in the activation of cell death. Differential expression of cuproptosis-related genes was detected in normal and malignant tissues. Cuproptosis-related genes were also linked to the regulation of oxidative stress, immune cell responses, and composition of tumor microenvironment. Activation of cuproptosis was associated with increased expression of redox-metabolism-regulating genes, such as ferredoxin 1 (FDX1), lipoic acid synthetase (LIAS), lipoyltransferase 1 (LIPT1), dihydrolipoamide dehydrogenase (DLD), drolipoamide S-acetyltransferase (DLAT), pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1), and pyruvate dehydrogenase E1 subunit beta (PDHB)). Accordingly, copper-activated network was suggested as an attractive target in cancer therapy. Mechanisms of cuproptosis and regulation of cuproptosis-related genes in different cancers and tumor microenvironment are discussed in this study. The analysis of current findings indicates that therapeutic regulation of copper signaling, and activation of cuproptosis-related targets may provide an effective tool for the improvement of immunotherapy regimens.
Collapse
Affiliation(s)
- Ruiwen Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Olga Sukocheva
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia.
| | - Edmund Tse
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia
| | - Margarita Neganova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yulia Aleksandrova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yufei Zheng
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Gu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Deyao Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - SabbaRao V Madhunapantula
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Xiaorong Zhu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Junqi Liu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ruitai Fan
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
40
|
Qian Z, Yu F. GADD45A: a key tumor suppressor in AML subtypes. Blood 2024; 144:6-7. [PMID: 38963670 DOI: 10.1182/blood.2024024685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024] Open
|
41
|
Yang F, Niu X, Zhou M, Li W. Development and validation of a novel disulfidptosis-related lncRNAs signature in patients with HPV-negative oral squamous cell carcinoma. Sci Rep 2024; 14:14436. [PMID: 38910181 PMCID: PMC11194273 DOI: 10.1038/s41598-024-65194-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: 04/13/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024] Open
Abstract
Disulfidptosis is a recently identified mode of regulated cell death. Regulating disulfidptosis in carcinoma is a promising therapeutic approach. Long non-coding RNAs (lncRNAs) have been reported to be related to the occurrence and development of many cancers. Disulfidptosis-related lncRNAs (DRLs) in HPV-negative oral squamous cell carcinoma (OSCC) have not been studied. Based on The Cancer Genome Atlas (TCGA) database, least absolute shrinkage selection operator (LASSO) analysis and Cox regression analysis were used to identify overall survival related DRLs and construct the signature. Kaplan-Meier, time-dependent receiver operating characteristics (ROC) and principal component analyses (PCA) were explored to demonstrate the prediction potential of the signature. Subgroup analysis stratified by different clinicopathological characteristics were conducted. Nomogram was established by DRLs signature and independent clinicopathological characteristics. The calibration plots were performed to reveal the accuracy of nomogram. Immune cell subset infiltration, immunotherapy response, drug sensitivity analysis, and tumor mutation burden (TMB) were conducted. Underlying functions and pathways were explored by Gene Set Enrichment Analysis (GSEA) analysis. Previous lncRNA signatures of OSCC were retrieved from PubMed for further validation. Gene expression omnibus (GEO) datasets (GSE41613 and GSE85446) were merged as an external validation for DRLs signature. Consensus clustering analysis of DRLs signature and experimental validation of DRLs were also explored. This research sheds light on the robust performance of DRLs signature in survival prediction, immune cell infiltration, immune escape, and immunotherapy of HPV-negative OSCC.
Collapse
Affiliation(s)
- Fan Yang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xinyu Niu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mingzhu Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Li
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|