1
|
Ashraf R, Adel M, Serya RAT, Ibrahim E, Haffez H, Soror S, Abouzid KAM. Design and synthesis of novel Hydroxamate and non-Hydroxamate HDAC inhibitors based on Chromone and Quinazolone scaffolds. Bioorg Chem 2025; 161:108514. [PMID: 40319810 DOI: 10.1016/j.bioorg.2025.108514] [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: 01/17/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
The development of selective histone deacetylase (HDAC) inhibitors represents an encouraging approach for cancer therapy. In this study, we report design, synthesis, and biological evaluation of hydroxamate, amidoxime, and carboxylic acid-based derivatives as novel HDAC inhibitors. The synthesized compounds were assessed for their inhibitory activity against multiple HDAC isoforms, particularly HDAC6, 7, and 8. Compounds 13, 16, 20, and 26 exhibited potent and selective inhibition of HDAC6. Compound 26 exhibited the most potent inhibitory activity against HDAC6, with an IC50 value of 70 nM. Additionally, compounds 17 and 23 demonstrated significant broad-spectrum antiproliferative activity across various cancer cell lines compared to other tested derivatives. Furthermore, compounds 17 and 23 showed promising total pan-HDAC inhibitory activity. Subsequent biological studies revealed that compounds 13, 16, 17, 20, 23, and 26 induced a combination of early and late apoptosis along with necrosis. In silico studies, including molecular docking and ADME predictions, were also conducted. Collectively, these findings highlight the potential of these compounds as promising candidates for the development of a novel class of selective HDAC6 inhibitors in the future.
Collapse
Affiliation(s)
- Rosaline Ashraf
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Mai Adel
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Rabah A T Serya
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Esraa Ibrahim
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt; Center of Scientific Excellence "Helwan Structural Biology Research, (HSBR)", Helwan University, 11795 Cairo, Egypt
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt; Center of Scientific Excellence "Helwan Structural Biology Research, (HSBR)", Helwan University, 11795 Cairo, Egypt
| | - Sameh Soror
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt
| | - Khaled A M Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt.
| |
Collapse
|
2
|
Kamala K, Ganapathy D, Sivaperumal P. Advancements in Cancer Therapy: Mycoviruses and Their Oncolytic Potential. Cell Biochem Biophys 2025; 83:1329-1332. [PMID: 39535660 DOI: 10.1007/s12013-024-01608-y] [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] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Recent advancements in cancer research focus on reducing treatment side effects while enhancing efficacy against medication resistance and tumor antigen detection. Genetic therapies utilizing microbes like bacteria, fungi, and viruses have garnered attention, with mycoviruses emerging as promising candidates. Particularly, the smallest fungal virus, Myco-phage, exhibits oncolytic properties by lysing cancer cells in the mouth, oral cavity, head, and neck without adverse effects. Genetically Modified Myco-phage (GmMP) adapts quickly to target cancer cells through cell membrane damage, inducing apoptosis and dendritic cell activation. Additionally, GmMP inhibits angiogenesis and modulates immune responses via CAR cells and immune checkpoints, potentially transforming cancer treatment paradigms with enhanced specificity and efficacy.
Collapse
Affiliation(s)
- Kannan Kamala
- Marine Microbial Research Lab, Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
- Centre for Marine and Aquatic Research (CMAR), Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 602105, India
| | - Dhanraj Ganapathy
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Pitchiah Sivaperumal
- Centre for Marine and Aquatic Research (CMAR), Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 602105, India.
- Marine Biomedical Research Lab & Environmental Toxicology Unit, Cellular and Molecular Research Centre, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India.
| |
Collapse
|
3
|
Xu G, Li L, Lv M, Li C, Yu J, Zeng X, Meng X, Yu G, Liu K, Cheng S, Luo H, Xu B. Discovery of novel 4-trifluoromethyl-2-anilinoquinoline derivatives as potential anti-cancer agents targeting SGK1. Mol Divers 2025; 29:1945-1965. [PMID: 39117890 DOI: 10.1007/s11030-024-10951-4] [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: 06/12/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Given the critical necessity for the development of more potent anti-cancer drugs, a series of novel compounds incorporating trifluoromethyl groups within the privileged 2-anilinoquinoline scaffold was designed, synthesized, and subjected to biological evaluation through a pharmacophore hybridization strategy. Upon evaluating the in vitro anti-cancer characteristics of the target compounds, it became clear that compound 8b, which contains a (4-(piperazin-1-yl)phenyl)amino substitution at the 2-position of the quinoline skeleton, displayed superior efficacy against four cancer cell lines by inducing apoptosis and cell cycle arrest. Following research conducted in a PC3 xenograft mouse model, it was found that compound 8b exhibited significant anti-cancer efficacy while demonstrating minimal toxicity. Additionally, the analysis of a 217-kinase panel pinpointed SGK1 as a potential target for this compound class with anti-cancer capabilities. This finding was further verified through molecular docking analysis and cellular thermal shift assays. To conclude, our results emphasize that compound 8b can be used as a lead compound for the development of anti-cancer drugs that target SGK1.
Collapse
Affiliation(s)
- Guangcan Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Lanlan Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Mengfan Lv
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Cheng Li
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Xiaoping Zeng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Xueling Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Gang Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Kun Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China.
| | - Bixue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China.
| |
Collapse
|
4
|
Kojja V, Kumar D, Kalavagunta PK, Bhukya B, Tangutur AD, Nayak PK. 2-(Diarylalkyl)aminobenzothiazole derivatives induce autophagy and apoptotic death through SIRT inhibition and P53 activation In MCF7 breast cancer cells. Comput Biol Chem 2025; 116:108395. [PMID: 39987744 DOI: 10.1016/j.compbiolchem.2025.108395] [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: 01/02/2025] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
Sirtuins (SIRTs) are multifunctional proteins that exhibit a wide range of substrate preferences and cellular localizations. They are reliant on NAD+ and are essential for the regulation of several cellular functions. The SIRT proteins play important role towards tumor survival and resistance mechanisms in tumor cells. Therefore, molecules targeting SIRT proteins gained significant recognition in cancer research. In this work, we explored the anticancer property, potential and mode of action of 2-(diarylalkyl)aminobenzothiazole derivatives on MCF7 human breast cancer cells. Our studies established that 2-(diarylalkyl)aminobenzothiazole derivatives 1-((6-chlorobenzo[d]thiazol-2-ylamino)(3,4-dichlorophenyl)methyl)naphthalen-2-ol (7ab) and 1-((6-chlorobenzo[d]thiazol-2-ylamino)(4-bromophenyl)methyl)naphthalen-2-ol (7ba) treatment in a dose dependent manner drastically lowered the cell proliferation in MCF7 cells and the IC50 values of 7ab and 7ba was found to be 11.4 µM and 9.6 µM at 24 hr in these cells. Docking and molecular dynamic simulation studies further revealed that 7ab and 7ba show significant binding with SIRT1 protein. Consistently, treatment with 7ab and 7ba reduced the expression levels of SIRT1 protein while increasing acetylation of p53, a known SIRT protein target in MCF-7 cells. We observed that SIRT1inhibition was associated with activation of p53, an essential protein for apoptotic cell death, in MCF-7 cell lines. Furthermore, 7ab and 7ba treatment induced LC3-II expression and vacuole formation in the cytoplasm leading to autophagic cell death. Our findings together reveal the plausible cellular targets and specificity of these new small molecules as SIRT inhibitors, which increase p53 acetylation and suppress the proliferation of MCF-7 human breast cancer cells by triggering autophagic and apoptotic cell death.
Collapse
Affiliation(s)
- Venkateswarlu Kojja
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Dinesh Kumar
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana State 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Praveen Kumar Kalavagunta
- Crop Protection Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana State 500007, India
| | - Bhima Bhukya
- Centre for Microbial and Fermentation Technology, Department of Microbiology, University College of Science, Osmania University, Hyderabad, Telangana State 500007, India
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana State 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| | - Prasanta Kumar Nayak
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
5
|
Mannan A, Mohan M, Singh TG. Revenge unraveling the fortress: Exploring anticancer drug resistance mechanisms in BC for enhanced therapeutic strategies. Crit Rev Oncol Hematol 2025; 210:104707. [PMID: 40122355 DOI: 10.1016/j.critrevonc.2025.104707] [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: 01/22/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Breast cancer (BC) is the most prevalent form of cancer in women worldwide and the main cause of cancer-related fatalities in females. BC can be classified into various types based on where cancer has begun to grow or spread, specific characteristics that influence how cancer behaves, and treatment choices. BC is multifaceted, and due to its diverse nature, the mechanisms involved are complex and have not yet been understood. Overexpression and expression of various factors involved in the functioning of mechanisms lead to abnormal changes, providing an environment supporting cancer cell growth. Understanding BC risk factors and early diagnosis through screening techniques like mammography and diagnostic techniques such as imaging and biopsies has advanced significantly. A wide range of treatment options, including surgery, radiation, chemotherapy, targeted treatments, and hormonal therapies, are now available. Daily advancements are being made in the clinical treatment of BC. Still, BC drug resistance cases remain highly prevalent and are currently one of the biggest problems faced by medical science. To increase response rates and possibly lengthen survival, there is a critical requirement for novel medicines with minimal sensitivity to overcome drug resistance. This review classifies different mechanisms that are involved in the development of BC and workable pharmacological targets and explains how they relate to the development of BC drug resistance. By concentrating on the mechanisms covered in this review, we can have a deep understanding of different mechanisms and learn innovative ways to develop novel therapeutics for the disease to combat medication resistance.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
6
|
He Y, Wang X. A comprehensive investigation of associations between cell death pathways and molecular and clinical features in pan-cancer. Clin Transl Oncol 2025; 27:2731-2749. [PMID: 39487950 DOI: 10.1007/s12094-024-03769-x] [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: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Regulated cell death (RCD) pathways play significant roles in tumorigenesis. However, systematic investigation into correlations between RCD and various molecular and clinical features, particularly anti-tumor immunity and immunotherapy response in pan-cancer remains lacking. METHODS Using the single-sample gene set enrichment analysis, we quantified the activities of six RCD pathways (apoptosis, autophagy, ferroptosis, cuproptosis, necroptosis, and pyroptosis) in each cancer specimen. Then, we explored associations of these six RCD pathways with tumor immunity, genomic instability, tumor phenotypes and clinical features, and responses to immunotherapy and targeted therapies in pan-cancer by statistical analyses. RESULTS Our results showed that the RCD (except autophagy) activities were oncogenic signatures, as evidenced by their hyperactivation in late stage or metastatic cancer patients, positive correlations with tumor proliferation, stemness, genomic instability and intratumor heterogeneity, and correlation with worse survival outcomes in cancer. In contrast, autophagy was a tumor suppressive signature as its associations with molecular and clinical features in cancer shows an opposite pattern compared to the other RCD pathways. Furthermore, heightened RCD (except cuproptosis) activities were correlated with increased sensitivity to immune checkpoint inhibitors. Additionally, elevated activities of pyroptosis, autophagy, cuproptosis and necroptosis were associated with increased drug sensitivity in a broad spectrum of anti-tumor targeted therapies, while the elevated activity of ferroptosis was correlated with decreased sensitivity to numerous targeted therapies. CONCLUSION RCD (except autophagy) activities correlate with unfavorable cancer prognosis, while the autophagy activity correlate with favorable clinical outcomes. RCD (except cuproptosis) activities are positive biomarkers for anti-tumor immunity and immunotherapy response.
Collapse
Affiliation(s)
- Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China.
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
7
|
Li F, Zhou J, Shi K, He Q, Diao W, Peng H, Liang G, Zhong C, Li W, Xu D. Ferroptosis and necroptosis may be involved in the formation and progression of hydrofluoric acid burn wounds: Results from an RNA-Seq analysis. Burns 2025; 51:107513. [PMID: 40327971 DOI: 10.1016/j.burns.2025.107513] [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: 09/14/2024] [Revised: 01/18/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Hydrofluoric acid (HF) burns have potentially serious consequences. The molecular mechanism of wound development is still unclear. This study aims to preliminarily explore the programmed cell death mode that may be involved in hydrofluoric acid burns by using transcriptome sequencing technology and to provide a theoretical basis for a new treatment approach for hydrofluoric acid burns. METHODS The rat model of hydrofluoric acid burn skin was constructed, and the differentially expressed genes after HF burn were screened by transcriptome sequencing technology. HE staining, TUNEL staining, immunohistochemistry, biochemical detection, and qRT-PCR were used to preliminarily verify the mode of cell death involved in hydrofluoric acid burn wounds. RESULTS The sequencing results suggest that the differential genes after HF burn were enriched in ferroptosis, apoptosis, and necroptosis pathways in cell growth and death aspects. HE staining confirmed HF burn wounds were progressively aggravated. The positive cells of TUNEL staining in the wound gradually increased. Compared with the normal group, the content of MDA in serum and skin tissue increased and the content of GSH decreased at 4, 8, 12, 24, and 48 hours after HF burn (P < 0.05). The level of serum Fe2 + in the HF burn group was higher than that in the normal group at 4 h, 8 h, and 12 h postburn (P < 0.05). The level of serum Fe2+ at 24 h and 48 h postburn was higher than that of the normal group, but the difference was not statistically significant. The content of Fe2+ in skin tissue increased and reached its peak at 12 h (P < 0.05). The serum calcium level decreased to its lowest level at 24 hours postburn (P < 0.05). Immunohistochemistry showed that the expressions of GPX4, FTH1, and Bcl-2 proteins in hydrofluoric acid burn wounds were down-regulated, while the expression of HO-1, Bax, RIPK1, and MLKL was increased (P < 0.05). RIPK3 expression was not significantly different. qRT-PCR showed that the expression of HO-1, FTH1, SLC39A14, SLC39A8, CYBB, ACSL4, Bax, RIPK1, MLKL, IL-1β, and IL-6 increased, while the expression of ACSL1, ACSL6, GPX4, and Bcl-2 decreased after hydrofluoric acid burn compared with the normal group (P < 0.05). The RIPK3 gene expression did not change significantly. CONCLUSIONS Ferroptosis and necroptosis are involved in the formation and progression of HF burn wounds. Early blocking of ferroptosis may be a potential therapeutic for blocking the progress of hydrofluoric acid burn wounds. Necroptosis involvment in the occurrence and development of hydrofluoric acid burn wounds may be a non-classical pathway.
Collapse
Affiliation(s)
- Fuying Li
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China; Department of Plastic and Burns Surgery, Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, China
| | - Jianda Zhou
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China
| | - Ke Shi
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China
| | - Quanyong He
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China
| | - Wuliang Diao
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Peng
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China
| | - Geao Liang
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China
| | - Chi Zhong
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China
| | - Wengjuan Li
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China
| | - Dan Xu
- Department of Burns and Plastic Surgery, the third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
8
|
Zuo X, Lei Y, Ou S, Yuan X, Shi P, Li Q, Xu Y. Integration of cuproptosis-related gene signatures in stomach adenocarcinoma: implications for prognostic prediction and therapeutic strategies in cancer drug resistance. Discov Oncol 2025; 16:885. [PMID: 40410601 PMCID: PMC12102457 DOI: 10.1007/s12672-025-02740-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 05/16/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is a prevalent and aggressive cancer, often diagnosed at later stages, which poses challenges for effective treatment. Despite advancements in cancer therapies, the phenomenon of tumor drug resistance remains a critical hurdle. Recent studies have highlighted cuproptosis, a copper-dependent regulated cell death process, as a potential mechanism in various cancers, including STAD. This study integrates cuproptosis-related gene signatures with clinical features to better predict prognosis and explore potential therapeutic targets, focusing on the role of cuproptosis in overcoming tumor resistance mechanisms. METHODS Using comprehensive datasets from TCGA-STAD (n = 375 tumor samples, 32 normal samples), GTEx (n = 211 normal gastric tissues), and GEO (GSE84437 and GSE29272), we analyzed the expression of genes associated with cuproptosis. We examined genetic alterations, immune infiltration, and constructed multivariate Cox regression models with clinicopathological covariates (age, gender, TNM stage, histological grade, residual tumor status) to assess the relationship between cuproptosis gene expression and patient survival outcomes, including overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI). Drug sensitivity analysis was performed using the Genomics of Drug Sensitivity in Cancer (GDSC) database. RESULTS Our analysis identified significant upregulation of several cuproptosis-related genes, including FDX1, which was correlated with improved prognosis and immune cell infiltration patterns. High expression of FDX1 was associated with better OS and DSS outcomes. Further genetic alterations, notably in CDKN2A, were frequent and linked to poor prognosis, highlighting the complexity of tumor drug resistance in STAD. Prognostic models incorporating FDX1, PDHA1, and LIAS expression stratified patients into distinct risk categories, emphasizing their potential as biomarkers for personalized therapeutic strategies. CONCLUSIONS This study underscores the importance of cuproptosis-related genes, particularly FDX1, in the prognosis and therapeutic response of STAD. By integrating molecular features with clinical data, we offer insights into the potential for overcoming drug resistance in cancer therapy. These findings lay the groundwork for future research into targeted treatments that modulate cuproptosis, offering a novel approach to tackling tumor progression and resistance in STAD.
Collapse
Affiliation(s)
- Xin Zuo
- Department of Gastroenterology, The Sixth People's Hospital of Chongqing, Chongqing, 400060, China
| | - Youchun Lei
- Department of Gastroenterology, The Sixth People's Hospital of Chongqing, Chongqing, 400060, China
| | - Shan Ou
- Department of Gastroenterology, The Sixth People's Hospital of Chongqing, Chongqing, 400060, China
| | - Xiu Yuan
- Department of Gastroenterology, The Sixth People's Hospital of Chongqing, Chongqing, 400060, China
| | - Peng Shi
- Department of Gastroenterology, The Sixth People's Hospital of Chongqing, Chongqing, 400060, China
| | - Qian Li
- Department of Gastroenterology, The Sixth People's Hospital of Chongqing, Chongqing, 400060, China
| | - Yun Xu
- Department of Gastroenterology, The Sixth People's Hospital of Chongqing, Chongqing, 400060, China.
| |
Collapse
|
9
|
Saadh MJ, Bishoyi AK, Rekha MM, Verma A, Nanda A, Panigrahi R, Verma R, Gabble BC. Dual roles of long non-coding RNAs in thyroid cancer: regulation of programmed cell death pathways. Med Oncol 2025; 42:217. [PMID: 40407962 DOI: 10.1007/s12032-025-02750-0] [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: 02/14/2025] [Accepted: 04/28/2025] [Indexed: 06/01/2025]
Abstract
Thyroid cancer (TC) represents the most common endocrine malignancy; however, the intricacies of its carcinogenesis pose significant challenges to therapeutic interventions. A comprehensive understanding of the molecular mechanisms that drive TC progression is crucial for the development of effective treatment strategies, especially considering the increasingly recognized role of non-coding RNAs (ncRNAs) in oncogenesis. Notwithstanding recent advancements, the regulatory functions of long non-coding RNAs (lncRNAs) and their interactions with microRNAs (miRNAs) in the context of TC are not yet fully elucidated. This review aims to address this knowledge deficiency by investigating the dual roles of lncRNAs in the pathogenesis of TC, specifically their regulation of programmed cell death (PCD) pathways. Current literature indicates that disrupted competitive endogenous RNA (ceRNA) networks are involved in drug resistance, epithelial-mesenchymal transition (EMT), as well as tumor proliferation, angiogenesis, invasion, and metastasis in TC. The basis of cancer therapy-induced tumor cell elimination is programmed cell death (PCD), which includes well-studied processes such as apoptosis, autophagy, and ferroptosis as well as novel pathways, such as cuproptosis, immunogenic cell death (ICD), and PANoptosis. Recent research has shown the critical function of long non-coding RNAs (lncRNAs) in modifying these several PCD pathways, impacting TC growth and therapy response. This review synthesizes evidence on how lncRNAs regulate PCD to influence TC progression and therapeutic outcomes. Additionally, we examine the clinical relevance of lncRNAs in TC, highlighting their potential as biomarkers for diagnosis and prognosis, therapeutic targets, and contributors to drug resistance, while emphasizing recent advancements in this field.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, 360003, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Ashish Verma
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Anima Nanda
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Rajashree Panigrahi
- Department of Microbiology, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab, 140307, India
| | - Baneen C Gabble
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
10
|
Wang P, Guo W, Liu S, Li S, Li J, Ding B, Yin F, Yang Y, Li X, Cao P, Ma C, Zhang W, Song Y, Geng Y, Liu L, Hu J, Hao J, Feng Y. Novel Pt@PCN-Cu-induced cuproptosis amplifies αPD-L1 immunotherapy in pancreatic ductal adenocarcinoma through mitochondrial HK2-mediated PD-L1 upregulation. J Exp Clin Cancer Res 2025; 44:149. [PMID: 40382627 DOI: 10.1186/s13046-025-03409-4] [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: 02/02/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Copper accumulation triggers mitochondrial-driven cell death, known as cuproptosis, offering a promising mechanism for targeted cancer therapy. Recent studies have highlighted the critical role of intratumoral copper levels in regulating the expression of programmed cell death ligand-1 (PD-L1), suggesting that copper-induced cuproptosis not only enhances cancer cell death but may also amplify the effects of anti-PD-L1 antibodies (αPD-L1). However, in tumors where monotherapy with αPD-L1 shows limited efficacy, particularly in pancreatic ductal adenocarcinoma (PDAC), the role of copper-induced cuproptosis in enhancing αPD-L1 treatment efficacy and its underlying mechanisms remain unclear. Meanwhile, inadequate tumor drug accumulation and glycolysis significantly restrict the efficacy of cuproptosis. To address these challenges, we have synthesized a novel nanozyme, Pt@PCN-Cu, designed to stabilize intracellular copper accumulation and effectively induce cuproptosis. Additionally, we aim to determine whether this strong induction of cuproptosis can synergize with αPD-L1 to enhance cancer therapy, ultimately paving the way for novel strategies to improve PDAC treatment. METHODS Pt@PCN-Cu was synthesized via a one-pot method, and its therapeutic potential was assessed in combination with αPD-L1 for the treatment of PDAC. Initially, the material's properties were characterized, and its efficient cellular uptake was confirmed. Anti-tumor efficacy was evaluated by inducing cuproptosis in PDAC cell lines and xenograft models. RNA sequencing (RNA-seq) was utilized to identify key regulators involved in the modulation of PD-L1 expression by cuproptosis. Lastly, the therapeutic efficacy of Pt@PCN-Cu combined with αPD-L1 was evaluated in vivo, focusing on tumor growth inhibition and immune modulation within the tumor microenvironment (TME). RESULTS Pt@PCN-Cu demonstrates excellent physicochemical properties and remarkable cascade catalytic activity, providing a solid foundation for further in vitro and in vivo studies. In vitro, Pt@PCN-Cu efficiently transports copper and induces cuproptosis primarily through mitochondrial dysfunction. Mechanistic studies show that Pt@PCN-Cu triggers the dissociation of hexokinase 2 (HK2) from mitochondria, leading to a reduction in HK2 activity. This decline in HK2 activity impairs glycolysis, a metabolic pathway essential for tumor energy metabolism, which in turn results in elevated PD-L1 levels. In vivo, Pt@PCN-Cu demonstrates excellent safety and accumulates at the tumor site in a subcutaneous PDAC mouse model, inducing cuproptosis. Moreover, the combination of Pt@PCN-Cu with αPD-L1 further enhanced its therapeutic efficacy and effectively reprogrammed the immunosuppressive TME. CONCLUSION This study presents strong evidence confirming the safety and therapeutic potential of Pt@PCN-Cu in PDAC treatment. Importantly, Pt@PCN-Cu not only induces cuproptosis but also significantly enhances antitumor efficacy in combination with αPD-L1 by regulating PD-L1 expression through HK2 modulation. These findings underscore a more effective and innovative approach for treating PDAC.
Collapse
Affiliation(s)
- Pengyu Wang
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Weihua Guo
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Shuyue Liu
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Shouyi Li
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jiaqi Li
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Bowen Ding
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Fengyi Yin
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yang Yang
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xingjiang Li
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Pei Cao
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Chaozhe Ma
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Wanying Zhang
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yidan Song
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yating Geng
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Lantao Liu
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China.
| | - Jing Hu
- School of Basic Medicine, Tianjin Medical University, Tianjin, 300070, China.
| | - Jihui Hao
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Yukuan Feng
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
11
|
Sun C, Gui J, Sheng Y, Huang L, Zhu X, Huang K. Specific signaling pathways mediated programmed cell death in tumor microenvironment and target therapies. Discov Oncol 2025; 16:776. [PMID: 40377777 PMCID: PMC12084487 DOI: 10.1007/s12672-025-02592-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
Increasing evidence has shown that programmed cell death (PCD) plays a crucial role in tumorigenesis and cancer progression. The components of PCD are complex and include various mechanisms such as apoptosis, necroptosis, alkaliptosis, oxeiptosis, and anoikis, all of which are interrelated in their functions and regulatory pathways. Given the significance of these processes, it is essential to conduct a comprehensive study on PCD to elucidate its multifaceted nature. Key signaling pathways, particularly the caspase signaling pathway, the RIPK1/RIPK3/MLKL pathway, and the mTOR signaling pathway, are pivotal in regulating PCD and influencing tumor progression. In this review, we briefly describe the generation mechanisms of different PCD components and focus on the regulatory mechanisms of these three major signaling pathways within the context of global PCD. Furthermore, we discuss various tumor therapeutic compounds that target different signaling axes of these pathways, which may provide novel strategies for effective tumor therapy and help improve patient outcomes in cancer treatment.
Collapse
Affiliation(s)
- Chengpeng Sun
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Jiawei Gui
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Yilei Sheng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Le Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China
| | - Xingen Zhu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Kai Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
12
|
Baldassarre G, L de la Serna I, Vallette FM. Death-ision: the link between cellular resilience and cancer resistance to treatments. Mol Cancer 2025; 24:144. [PMID: 40375296 PMCID: PMC12080166 DOI: 10.1186/s12943-025-02339-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025] Open
Abstract
One of the key challenges in defeating advanced tumors is the ability of cancer cells to evade the selective pressure imposed by chemotherapy, targeted therapies, immunotherapy and cellular therapies. Both genetic and epigenetic alterations contribute to the development of resistance, allowing cancer cells to survive initially effective treatments. In this narration, we explore how genetic and epigenetic regulatory mechanisms influence the state of tumor cells and their responsiveness to different therapeutic strategies. We further propose that an altered balance between cell growth and cell death is a fundamental driver of drug resistance. Cell death programs exist in various forms, shaped by cell type, triggering factors, and microenvironmental conditions. These processes are governed by temporal and spatial constraints and appear to be more heterogeneous than previously understood. To capture the intricate interplay between death-inducing signals and survival mechanisms, we introduce the concept of Death-ision. This framework highlights the dynamic nature of cell death regulation, determining whether specific cancer cell clones evade or succumb to therapy. Building on this understanding offers promising strategies to counteract resistant clones and enhance therapeutic efficacy. For instance, combining DNMT inhibitors with immune checkpoint blockade may counteract YAP1-driven resistance or the use of transcriptional CDK inhibitors could prevent or overcome chemotherapy resistance. Death-ision aims to provide a deeper understanding of the diversity and evolution of cell death programs, not only at diagnosis but also throughout disease progression and treatment adaptation.
Collapse
Affiliation(s)
- Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, 33081, Italy.
| | - Ivana L de la Serna
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| | - François M Vallette
- Centre de Recherche en Cancérologie et Immunologie Intégrées Nantes Angers (CRCI2 NA), INSERM UMR1307/CNRS UMR 6075/Nantes Université/Univ. Angers. Nantes, 44007, Nantes, France.
- Institut de Cancérologie de L'Ouest (ICO), 44085, Saint-Herblain, France.
| |
Collapse
|
13
|
Li Y, Yu Y, Hu S, Li S. Identification of programmed cell death-related genes and construction of a prognostic model in oral squamous cell carcinoma using single-cell and transcriptome analysis. Discov Oncol 2025; 16:713. [PMID: 40346375 PMCID: PMC12064537 DOI: 10.1007/s12672-025-02520-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is characterized by poor prognosis and high mortality. Understanding programmed cell death-related genes could provide valuable insights into disease progression and treatment strategies. METHODS RNA-sequencing data from 341 OSCC tumor tissues and 31 healthy samples were analyzed from TCGA database, with validation using 76 samples from GSE41613. Single-cell RNA sequencing data was obtained from GSE172577 (6 OSCC samples). Differentially expressed genes (DEGs) were identified and intersected with 1,254 programmed cell death-related genes. A protein-protein interaction network was constructed, and key modules were identified. Univariate Cox, LASSO, and multivariate Cox regression analyses were performed to build a prognostic model. Model performance was evaluated using Kaplan-Meier analysis, ROC curves, and nomogram validation. RESULTS The study identified 200 candidate genes from the intersection of DEGs and programmed cell death-related genes, which were further refined to 57 hub genes through PPI network analysis. A prognostic signature consisting of five genes (MET, GSDMB, KIT, PRKAG3, and CDKN2A) was established and validated. The model demonstrated good predictive performance in both training and validation cohorts (AUC > 0.6 for 1-, 2-, and 3-year survival). Single-cell analysis revealed that prognostic genes were predominantly expressed in stromal and epithelial cells. Cell communication analysis indicated strong interactions between stromal and epithelial cells. CONCLUSIONS This study developed and validated a novel five-gene prognostic signature for OSCC based on programmed cell death-related genes. The model shows promising clinical application potential for risk stratification and personalized treatment of OSCC patients.
Collapse
Affiliation(s)
- Yongheng Li
- Department of Stomatology, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Harbin, 150001, Heilongjiang Province, China.
| | - Yang Yu
- Department of Stomatology, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Harbin, 150001, Heilongjiang Province, China
| | - Shaonan Hu
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Simin Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| |
Collapse
|
14
|
Yan Z, Zheng G, Zou J, Zou X, Chai K, Zhang G. Cathepsin B in urological tumors: unraveling its role and therapeutic potential. Discov Oncol 2025; 16:707. [PMID: 40343561 PMCID: PMC12064525 DOI: 10.1007/s12672-025-02552-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 05/02/2025] [Indexed: 05/11/2025] Open
Abstract
Cathepsin B(CTSB) is a key protease within the lysosomal protease family and is recognized as a tumor-promoting factor that exerts a substantial impact on cancer progression. It plays a critical role in the initiation, proliferation, metastasis, and angiogenesis of cancer, significantly advancing the disease. This review offers a concise overview of the structure and biological functions of CTSB, clarifying its relationship with cancer and the role it plays in the disease's progression. Additionally, we discuss the association between CTSB and several common malignant tumors of the urinary system, highlighting its potential role and clinical significance within these tumors, as well as the challenges that remain.
Collapse
Affiliation(s)
- Zhaojie Yan
- First Clinical College, Gannan Medical University, Ganzhou, 341000, China
| | - Guansong Zheng
- First Clinical College, Gannan Medical University, Ganzhou, 341000, China
| | - Junrong Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
- Institute of Urology, Gannan Medical University, Ganzhou, 341000, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, 341000, China
| | - Xiaofeng Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Keqiang Chai
- Department of Urology, Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, 730900, China.
| | - Guoxi Zhang
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
15
|
Lin Z, Li W, Xu Y, Liu H, Zhang Y, Li R, Zhao W, Guan Y, Zhang X. Identification of regulatory cell death-related genes during MASH progression using bioinformatics analysis and machine learning strategies. Front Immunol 2025; 16:1542524. [PMID: 40406118 PMCID: PMC12094957 DOI: 10.3389/fimmu.2025.1542524] [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: 12/10/2024] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Background Metabolic dysfunction-associated steatohepatitis (MASH) is becoming increasingly prevalent. Regulated cell death (RCD) has emerged as a significant disease phenotype and may act as a marker for liver fibrosis. The present study aimed to investigate the regulation of RCD-related genes in MASH to elucidate the role of RCD in the progression of MASH. Methods The gene expression profiles from the GSE130970 and GSE49541 datasets were retrieved from the Gene Expression Omnibus (GEO) database for analysis. A total of 101 combinations of 10 machine learning algorithms were employed to screen for characteristic RCD-related differentially expressed genes (DEGs) that reflect the progression of MASH. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted to explore the enrichment pathways and functions of the feature genes. we performed cell classification analysis to investigate immune cell infiltration. Consensus cluster analysis was performed to identify MASH subtypes associated with RCD. The GSE89632 dataset was utilized to analyze the correlation of characteristic genes with clinical features of MASH. The DGIdb database was employed to screen for potential therapeutic drugs and compounds targeting the feature genes. In addition, we established mouse liver fibrosis models induced by methionine-choline-deficient (MCD) diet or CCl4 treatment, and further validated the expression of characteristic genes through quantitative real-time PCR (q-PCR). Lastly, we knocked down EPHA3 in LX2 cells to explore its effect on TGFb-induced activation of LX2 cells. Results This study discovered a total of 11 RCD-associated DEGs, which predicted the progression of MASH. Advanced MASH has higher levels of immune cell infiltration and is significantly correlated with the RCD-related DEGs expression. MASH can be classified into two subtypes, cluster 1 and cluster 2, based on these feature genes. Compared with cluster 1, cluster 2 has highly expressed RCD-related DEGs, shows an increase in the degree of fibrosis. Furthermore, We discovered that the expression levels of feature genes were positively correlated with AST and ALT levels. Subsequently, We also evaluated the expression of these 11 feature genes in the liver tissues of mice with fibrosis induced by MCD or CCl4, and the results suggested that these genes may be involved in the development of fibrosis. WB results showed that the protein level of EPHA3 significantly increased in both mouse models of liver fibrosis. In vitro, we observed that knocking down EPHA3 in LX2 cells significantly inhibited the activation of the TGF-β/Smad3 signaling pathway. Conclusion Our study sheds light on the fact that RCD contribute to the progression of MASH, high lighting potential therapeutic targets for treating this disease.
Collapse
Affiliation(s)
- Zhiqiang Lin
- Health Science Center, East China Normal University, Shanghai, China
| | - Weiyi Li
- Department of Nephrology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yuan Xu
- Health Science Center, East China Normal University, Shanghai, China
| | - Hangchi Liu
- Health Science Center, East China Normal University, Shanghai, China
| | - Yufei Zhang
- Health Science Center, East China Normal University, Shanghai, China
| | - Ruifen Li
- Health Science Center, East China Normal University, Shanghai, China
| | - Wenqian Zhao
- Health Science Center, East China Normal University, Shanghai, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyan Zhang
- Health Science Center, East China Normal University, Shanghai, China
| |
Collapse
|
16
|
Yu H, Xue T, Mao X. Chinese herbal extracts mediated programmed cell death in cancer and inflammation therapy. J Leukoc Biol 2025; 117:qiaf051. [PMID: 40313183 DOI: 10.1093/jleuko/qiaf051] [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: 08/23/2024] [Revised: 12/13/2024] [Accepted: 04/30/2025] [Indexed: 05/03/2025] Open
Abstract
Programmed cell death is a common phenomenon in the development of organisms. It is an active and orderly mode of cell death determined by genes. Programmed cell death is usually classified into 3 different types according to the cell morphological changes, stimulus, and biochemical pathways involved, namely, apoptosis, programmed necrosis, and autophagy. Chinese herbal extracts, mainly obtained from traditional Chinese medicine and their primary plants through the physicochemical extraction and separation process, are concentrated with 1 or more effective ingredients from the herbal materials. Recently, studies focused on the influence of traditional Chinese medicine on programmed cell death are increasing, involving the protection of the nervous system and cardio-cerebrovascular system, the prevention of gastrointestinal and immune function damage, the treatment against tumors, and so on. This review mainly focuses on the effects of Chinese herbal extracts on various types of programmed cell death. In addition, the therapeutic approaches and prospects of CHEs are also discussed. Although there are promising clinical applications of Chinese herbal extracts, some challenges are still waiting to be overcome by further research for the wider use of Chinese herbal extracts in clinical practice.
Collapse
Affiliation(s)
- Haihong Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078
| | - Tingmao Xue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078
- Department of Pharmaceutical Sciences, Faculty of Health Science, University of Macau, Macao SAR, 999078, China
| |
Collapse
|
17
|
Shang N, Wang J, Liu Z, Wang Y, Zhang D, Liu H, Zhang Y, Dai G, Guan X. Integrated Analysis of Disulfidptosis-Related Genes Identifies CD2AP as a Potential Therapeutic Target for Hepatocellular Carcinoma. Int J Mol Sci 2025; 26:4454. [PMID: 40362690 PMCID: PMC12072785 DOI: 10.3390/ijms26094454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly cancer with limited treatment options for patients at advanced stages. It is urgent to develop reliable prognostic risk models and identify more biomarkers to improve the clinical outcomes of patients with HCC. Disulfidptosis is a newly discovered form of regulated cell death (RCD), and research on the comprehensive roles of disulfidptosis-related genes (DRGs) in HCC prognosis and development remains limited. In this paper, we systematically analyzed the expression levels and prognostic profiles of 26 DRGs in HCC samples from The Cancer Genome Atlas (TCGA) cohort and developed a prognostic risk model using seven hub DRGs. The independent prognostic value of the risk model was further validated in the external cohort. The overall survival of patients with HCC in the low-risk group was significantly longer than that of those in the high-risk group. Subsequently, the protein level of CD2-associated protein (CD2AP) was found to be highly expressed in HCC clinical tissues and associated with the severity of HCC. In vitro experiments demonstrated that the down-regulation of CD2AP attenuated the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) abilities of HCC cells. Taken together, our study revealed that the DRG CD2AP may serve as a potential biomarker for HCC and offer support for prognosis prediction of patients with HCC.
Collapse
Affiliation(s)
- Ning Shang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Jianwei Wang
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450001, China;
| | - Zihan Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Yake Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Di Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Huanfei Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Yaqing Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Guifu Dai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| | - Xiaowen Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (N.S.); (Z.L.); (Y.W.); (D.Z.); (H.L.); (Y.Z.)
| |
Collapse
|
18
|
An Y, Liu W, Deng Y, Huang W, Huang J. SLC7A11-HSPB1 Axis: A Novel Mechanism for Hepatocellular Carcinoma Progression and Ferroptosis Regulation. Biomed J 2025:100869. [PMID: 40339903 DOI: 10.1016/j.bj.2025.100869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/27/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND SLC7A11, a plasma membrane protein, has been implicated as an oncogene in various cancers, including hepatocellular carcinoma (HCC). Its role in HCC pathogenesis, particularly in relation to ferroptosis, is not well understood. This study aims to investigate the function of SLC7A11 with ferroptosis and its interaction in development of HCC. METHODS AND MATERIAL Clinical HCC tissue samples were used to analyze the expression of SLC7A11 by RT-PCR. The impact of SLC7A11 on HCC cell viability, proliferation, and migration was assessed by CCK-8, AlamarBlue and Transwell. Protein-protein interactions were explored using co-immunoprecipitation and immunofluorescence. The effect of SLC7A11 on ferroptosis was evaluated by iron levels, ROS, and GSH. The impact of sorafenib and doxorubicin (DOX) on HCC cells was analyzed using cell viability assay. RESULTS SLC7A11 was found to be highly expressed in HCC tissues and was correlated with tumor size and poor prognosis. Overexpression of SLC7A11 in HCC cells promoted cell viability, proliferation, and migration. Additionally, SLC7A11 overexpression mitigated erastin-induced ferroptosis, as evidenced by decreased ROS levels and increased GSH levels. We also discovered that SLC7A11 interacted with HSPB1. HSPB1 inhibited erastin-induced ferroptosis. Furthermore, a portion of the cell death induced by sorafenib and DOX is attributed to ferroptosis, with HSPB1 and SLC7A11 inhibiting the death induced by the two drugs, respectively. CONCLUSIONS SLC7A11 plays a significant role in HCC progression by inhibiting ferroptosis, and its interaction with HSPB1 is a critical pathway in this process. Targeting the SLC7A11-HSPB1 axis may provide a novel therapeutic strategy for HCC treatment, highlighting the importance of understanding the mechanisms of ferroptosis in cancer cells.
Collapse
Affiliation(s)
- Yan An
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China; Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200050, China
| | - Weilong Liu
- Institute of Hepatology, National clinical research center for infectious diseases, Guangdong Key Lab for Diagnosis &Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
19
|
Lin Y, Zhang Y, Huang T, Chen J, Li G, Zhang B, Xu L, Wang K, He H, Chen H, Liu D, Guo S, He X, Lan P. Arginine Deprivation Induces Quiescence and Confers Vulnerability to Ferroptosis in Colorectal Cancer. Cancer Res 2025; 85:1663-1679. [PMID: 39992728 PMCID: PMC12046318 DOI: 10.1158/0008-5472.can-24-1940] [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: 06/09/2024] [Revised: 11/21/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
Metabolic reprogramming is a hallmark of cancer. Rewiring of amino acid metabolic processes provides the basis for amino acid deprivation therapies. In this study, we found that arginine biosynthesis is limited in colorectal cancer because of the deficiency of ornithine transcarbamylase. Accordingly, colorectal cancer cells met the demand for arginine by increasing external uptake. The addiction to environmental arginine resulted in the susceptibility of colorectal cancer to arginine deprivation, which dramatically decreased proliferation in colorectal cancer cells and promoted these cells to enter a reversible quiescence state. Arginine deprivation induced quiescence by activating the AMPK-p53-p21 pathway. RNA sequencing data indicated that colorectal cancer cells may be vulnerable to ferroptosis during arginine deprivation and the combination of ferroptosis inducers and arginine deprivation strongly impeded tumor growth in vivo. These findings suggest that dietary modification combined with ferroptosis induction could be a potential therapeutic strategy for colorectal cancer. Significance: Colorectal cancer dependency on arginine uptake creates a metabolic vulnerability to arginine deficiency that causes cell cycle arrest and ferroptosis sensitivity, highlighting arginine deprivation plus ferroptosis induction as a promising treatment.
Collapse
Affiliation(s)
- Yanyun Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanhong Zhang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianze Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junguo Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guanman Li
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bin Zhang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Xu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Anaesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Danling Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuang Guo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaosheng He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping Lan
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| |
Collapse
|
20
|
Mao S, Li Q, Yang Y, Liu Z, Zhang L. Potential Crosstalk Between ANXA1+ Epithelial Cells and FABP4+ TAM Cells of Ferroptosis-Related Molecular Clusters Promotes an Immunosuppressive Microenvironment in Non-Small Cell Lung Cancer. Mol Carcinog 2025; 64:936-950. [PMID: 40040274 DOI: 10.1002/mc.23899] [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: 09/25/2024] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 03/06/2025]
Abstract
The tumor microenvironment (TME) affects tumor initiation, invasion, metastasis, and therapies. Recently, increasing evidence has demonstrated that ferroptosis plays important regulatory roles in tumourigenesis and progression. It is unclear how ferroptosis affects non-small cell lung cancer (NSCLC) progression by remodeling the TME. In this study, the single-cell RNA sequencing (scRNA-seq) data (85,562 cells, n = 18) were employed to reveal the heterogeneity of ferroptosis activation in NSCLC, and identified six ferroptosis-related molecular clusters. We found that ANXA1+ epithelial and FABP4 + TAM subpopulations were key factors in lung cancer progression and TME remodeling. In addition, the cell-cell communication analysis showed that ANXA1-FPR2/FPR1 receptor-ligand pair contributed to the formation of an immunosuppressive TME. Furthermore, we established a novel signature based on ferroptosis-related molecular clusters, and the risk score model may predict survival and response to immunotherapy. We also found that compared with responder, the expression of ANXA1 and FABP4 is higher in progressor, which indicating a higher expression of ANXA1 and FABP4 was associated with a worse response to immunotherapy. Therefore, we concluded that the molecular clusters associated with ferroptosis served as potential prognostic markers and therapeutic targets for NSCLC patients.
Collapse
Affiliation(s)
- Shengqiang Mao
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingyan Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ying Yang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhiqiang Liu
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
21
|
Kurmangaliyeva S, Baktikulova K, Tkachenko V, Seitkhanova B, Shapambayev N, Rakhimzhanova F, Almagambetova A, Kurmangaliyev K. An Overview of Hexavalent Chromium-Induced Necroptosis, Pyroptosis, and Ferroptosis. Biol Trace Elem Res 2025; 203:2619-2635. [PMID: 39287767 DOI: 10.1007/s12011-024-04376-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Heavy metals are common environmental industrial pollutants. Due to anthropogenic activity, chromium, especially its hexavalent form [Cr(VI)], is a widespread environmental contaminant that poses a threat to human health. In this review paper, we summarize the currently reported molecular mechanisms involved in chromium toxicity with a focus on the induction of pro-inflammatory non-apoptotic cell death pathways such as necroptosis, pyroptosis, and ferroptosis. The review highlights the ability of chromium to induce necroptosis, pyroptosis, and ferroptosis revealing the signaling pathways involved. Cr(VI) can induce RIPK1/RIPK3-dependent necroptosis both in vitro and in vivo. Chromium toxicity is associated with pyroptotic NLRP3 inflammasome/caspase-1/gasdermin D-dependent secretion of IL-1β and IL-18. Furthermore, this review emphasizes the role of redox imbalance and intracellular iron accumulation in Cr(VI)-induced ferroptosis. Of note, the crosstalk between the investigated lethal subroutines in chromium-induced toxicity is primarily mediated by reactive oxygen species (ROS), which are suggested to act as a rheostat determining the cell death pathway in cells exposed to chromium. The current study provides novel insights into the pro-inflammatory effects of chromium, since necroptosis, pyroptosis, and ferroptosis affect inflammation owing to their immunogenic properties linked primarily with damage-associated molecular patterns. Inhibition of these non-apoptotic lethal subroutines can be considered a therapeutic strategy to reduce the toxicity of heavy metals, including chromium.
Collapse
Affiliation(s)
- Saulesh Kurmangaliyeva
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kristina Baktikulova
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan.
| | - Viktoriya Tkachenko
- State Institution "Republican Scientific and Practical Centre of Sports, " 8 Narochanskaya St, Minsk, Republic of Belarus
| | - Bibigul Seitkhanova
- Department of Microbiology, Virology and Immunology, South Kazakhstan Medical Academy, Al-Farabi Sq, Shymkent, Republic of Kazakhstan
| | - Nasriddin Shapambayev
- Department of General Practitioner - 1, Khoja Akhmet Yasawi International Kazakh-Turkish University, 7/7 Baitursynov St, Shymkent, Republic of Kazakhstan
| | - Farida Rakhimzhanova
- Department of Microbiology, NCJSC "Semey Medical University, " 103 Abay St, Semey, Republic of Kazakhstan
| | - Altyn Almagambetova
- Department of Phthisiology and Dermatovenerology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kairat Kurmangaliyev
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| |
Collapse
|
22
|
Chen Y, Zhang J, Xu H. Recent developments in cuproptosis of glioblastoma. Pathol Res Pract 2025; 269:155939. [PMID: 40164043 DOI: 10.1016/j.prp.2025.155939] [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: 12/18/2024] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Glioblastoma (GBM) is the most malignant tumor within the central nervous system, attributed to its high-grade malignancy, propensity for recurrence, refractoriness to conventional therapeutic modalities, and the suboptimal efficacy of current targeted therapies. Hence, there is an urgent need to identify more efficacious molecular targets for the therapeutic intervention of GBM. The regulated cell death (RCD) has specific signaling factors and signaling pathways. Hence, targeting RCD is considered to be one of the effective targeted therapies for GBM. At present, cuproptosis is a novel form of RCD, characterized by a distinct molecular mechanism that differentiates it from apoptosis, pyroptosis, necroptosis, and ferroptosis. It is characterized by its principal mechanisms, which include copper dependency, the accumulation of acylated proteins, and the reduction of Fe-S cluster-containing proteins. These processes collectively induce proteotoxic stress, culminating in cell death. In previous studies, copper-ionized formulations have demonstrated cytotoxic effects on gliomas. Thus, the key factors of cuproptosis may be able to serve as a new target for GBM treatment. This review delves into several pivotal aspects, including the discovery of cuproptosis, the impact of copper homeostasis on tumorigenesis, the role of cuproptosis in GBM, and its potential as a therapeutic target in molecular targeted therapy for GBM. Hence, this article could reveal novel strategies for GBM treatment.
Collapse
Affiliation(s)
- Yajia Chen
- Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China
| | - Jingxian Zhang
- Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China
| | - Hongwu Xu
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Wandao Rd, 78, Wanjiang Subdistrict, Dongguan, Guangdong Province 523059, China; Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China.
| |
Collapse
|
23
|
Tang Z, Zhang Z, Zhao J, Zhang F, Zhang Y, Wen Y, Li M, Sun J, Shi L, Chen W, Li Z, Guo Z, Liu Y. Integrated analysis of multiple programmed cell death-related prognostic genes and functional validation of apoptosis-related genes in osteosarcoma. Int J Biol Macromol 2025; 307:142113. [PMID: 40089239 DOI: 10.1016/j.ijbiomac.2025.142113] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
Osteosarcoma (OS) is one of the most prevalent bone malignancies with a poor prognosis. Various types of programmed cell death patterns can influence cancer progression and response to treatment. We aimed to integrate different molecular characteristics of cell death for risk stratification and personalized therapy. First, we obtained transcriptomic, single-cell transcriptomic, and clinical information from the TARGET-OS and GEO databases as well as analyzed genes in fourteen cell death patterns to establish the cell death index (CDI) signature. A nomogram constructed from the CDI calculated from seven genes in combination with metastasis could effectively predict the prognosis of OS patients. Subsequently, the prognostic value and immune characteristics in CDI-defined subgroups were analyzed. A construct nomogram model was also constructed with clinical information. Notably, immunohistochemistry confirmed that the expression of GALNT14, a core gene in CDI model, correlated with poor survival. Deficiency of the highly expressed prognostic gene GALNT14 significantly repressed OS progression and OS cell proliferation by promoting apoptosis. We subsequently demonstrated that Bortezomib, a targeted inhibitor of GALNT14, can be used to enhance chemosensitivity. Finally, it was further elucidated that Bortezomib reduces MT2A glycosylation and improves its stability to promote apoptosis in OS cells by inhibiting GALNT14 expression. In summary, integration of multiple cell death genes may improve the ability to stratify risk in patients with OS, and targeting GALNT14 with Bortezomib improves chemotherapy sensitivity and induces apoptosis.
Collapse
Affiliation(s)
- Zhen Tang
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China; Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Zhi Zhang
- Department of Orthopedic Surgery, Guyuan People's Hospital, Ningxia, China
| | - Jungang Zhao
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Fan Zhang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Yiran Zhang
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yanhua Wen
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Miaozhen Li
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jin Sun
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Lei Shi
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Wentian Chen
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China.
| | - Zheng Guo
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China.
| | - Yingxiang Liu
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China.
| |
Collapse
|
24
|
Zhang S, Bi Y, Xiang K, Tang Y. P2X 7 Receptor Facilitates Cardiomyocyte Autophagy After Myocardial Infarction via Nox4/PERK/ATF4 Signaling Pathway. Cell Biochem Funct 2025; 43:e70078. [PMID: 40289672 DOI: 10.1002/cbf.70078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Myocardial infarction (MI) represents a critical cardiovascular emergency, standing as a leading cause of global mortality. ATP, a typical damage-associated molecular pattern, is stored in cells at high concentrations. Upon cellular injury, hypoxia, or necrosis, substantial quantities of ATP efflux into the extracellular space, activating P2X7 receptors, thereby initiating multiple signaling cascades. In vivo studies demonstrated coordinated upregulation of P2X7 and autophagy-related proteins in the infarcted border zone. Transcriptome sequencing revealed Nox4 overexpression in the myocardial tissue post-infarction; furthermore, administration of the P2X7 receptor antagonist A740003 effectively reduced both autophagy-related protein levels and Nox4 expression. In vitro experiments indicated that hypoxia induced upregulation of Nox4, p-PERK/PERK, ATF4, Beclin-1, and ATG5 in cardiomyocytes, A740003 could inhibit the expression of these proteins, while overexpression of Nox4 counteracted this effect. Collectively, our findings indicated that the P2X7 receptor expression was elevated in the infarcted border zone following MI and implicated its role in excessive autophagy induced by hypoxia in cardiomyocytes-at least partially through the Nox4/PERK/ATF4 pathway, thereby exacerbating myocardial injury following MI.
Collapse
Affiliation(s)
- Shuhong Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, People's Republic of China
| | - Yingying Bi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, People's Republic of China
| | - Kaili Xiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, People's Republic of China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
25
|
Yan W, Xiang S, Feng J, Zu X. Role of ubiquitin-specific proteases in programmed cell death of breast cancer cells. Genes Dis 2025; 12:101341. [PMID: 40083330 PMCID: PMC11904532 DOI: 10.1016/j.gendis.2024.101341] [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: 01/20/2024] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 03/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignant tumor and the leading cause of cancer-related deaths among women worldwide. Great progress has been recently achieved in controlling breast cancer; however, mortality from breast cancer remains a substantial challenge, and new treatment mechanisms are being actively sought. Programmed cell death (PCD) is associated with the progression and treatment of many types of human cancers. PCD can be divided into multiple pathways including autophagy, apoptosis, mitotic catastrophe, necroptosis, ferroptosis, pyroptosis, and anoikis. Ubiquitination is a post-translational modification process in which ubiquitin, a 76-amino acid protein, is coupled to the lysine residues of other proteins. Ubiquitination is involved in many physiological events and promotes cancer development and progression. This review elaborates the role of ubiquitin-specific protease (USP) in programmed cell death, which is common in breast cancer cells, and lays the foundation for tumor diagnosis and targeted therapy.
Collapse
Affiliation(s)
| | | | - Jianbo Feng
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| | - Xuyu Zu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| |
Collapse
|
26
|
Ma W, Wang Q, Guo L, Ju X. The molecular mechanisms, roles, and potential applications of PANoptosis in cancer treatment. Front Immunol 2025; 16:1550800. [PMID: 40364845 PMCID: PMC12069359 DOI: 10.3389/fimmu.2025.1550800] [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: 12/24/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
PANoptosis, a newly identified form of programmed cell death regulated by the panoptosome complex, exhibits key characteristics of apoptosis, pyroptosis and necroptosis. It exerts a substantial influence on the initiation and progression of a spectrum of diseases, particularly in cancer, where its impact is increasingly being recognized. PANoptosis is closely related to tumorigenesis, carcinogenesis, metastasis, chemotherapy resistance, as well as the prediction of therapeutic responses and prognosis in cancer patients. In this review, we first review the discovery of PANoptosis and systematically analyze the composition of the panoptosome. Subsequently, we examine the role of PANoptosis in various types of cancer, encompassing its function within the tumor microenvironment, its role in tumor drug resistance, and its predictive role in cancer prognosis. Ultimately, we delve into strategies for targeting PANoptosis in cancer therapy, including targeting various molecules in the PANoptosis pathway, such as ZBP1, RIPK1, RIPK3, Caspases and other novel strategies like nanoinducers and viral vectors. This review aims to provide references and assistance for the research and application of PANoptosis in cancer treatment.
Collapse
Affiliation(s)
- Wenyuan Ma
- Department of Pathology, The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang Clinical Medical College of Jiangsu University, Danyang, Jiangsu, China
| | - Qiang Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Lanfang Guo
- Department of Clinical Laboratory Medicine, The Fourth People’s Hospital of Jiangsu University, Zhenjiang, Zhenjiang, Jiangsu, China
| | - Xiaoli Ju
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
27
|
Li P, Zhang J, Shao T, Jiang J, Tang X, Yang J, Li J, Fang B, Huang Z, Fang H, Wang H, Hu W, Peng B, Bai H, Li L. NIR-II Photosensitizer-Based Nanoparticles Defunctionalizing Mitochondria to Overcome Tumor Self-Defense by Promoting Heat Shock Protein 40. ACS NANO 2025; 19:15751-15766. [PMID: 40241294 DOI: 10.1021/acsnano.4c18937] [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: 04/18/2025]
Abstract
Inherent self-defense pathways within malignant tumors include the action of heat shock proteins (HSPs) and often impede photothermal therapy efficacy. Interestingly, HSP40 inhibits glycolysis and disrupts mitochondrial function to overcome tumor self-defense mechanisms and exhibits a tumor-suppressive effect. Reactive oxygen species (ROS), especially hydroxyl radicals, generated by type-I photodynamic therapy inhibit adenosine triphosphate (ATP) production and lead to ATP-independent HSP40 overexpression during heat stress. However, the regulatory mechanisms linking heat and hydroxyl radicals to induce HSP40 expression remain unclear. Therefore, it is imperative to elucidate the underlying mechanism governing the induction of HSP40 expression during heat stress and explore its potential as a promising therapeutic strategy against tumor development. By strategically modifying the aza-BODIPY structure to precisely distribute the excited-state energy, we have demonstrated that HSP40 specific expression is correlated with the proportion of heat to hydroxyl radicals rather than their individual levels. This orchestrated NIR-II photosensitizer-based nanoparticles reduced tumor glycolysis and disrupted ATP production, driving cell apoptosis and amplifying the efficacy of photothermal therapy. Silencing and compensation of HSPs under heat and ROS stress represent a promising and effective strategy for overcoming tumor self-defense mechanisms in cancer therapy.
Collapse
Affiliation(s)
- Panpan Li
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Jiaxin Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Tao Shao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Jiamin Jiang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Xiao Tang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Jiaqi Yang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Jintao Li
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Bin Fang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Ze Huang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen 361102, China
| | - Haixiao Fang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen 361102, China
| | - Hui Wang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen 361102, China
| | - Wenbo Hu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Bo Peng
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Hua Bai
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Lin Li
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen 361102, China
| |
Collapse
|
28
|
Li J, Jiang Y, Nong S, Liang L, Chen L, Gong Q. Development of a machine learning-derived programmed cell death index for prognostic prediction and immune insights in colorectal cancer. Discov Oncol 2025; 16:608. [PMID: 40274671 PMCID: PMC12021754 DOI: 10.1007/s12672-025-02323-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Colorectal cancer (CRC) is a major contributor to cancer-related mortality worldwide, emphasizing the need for improved prognostic tools and therapeutic strategies. Programmed cell death, encompassing diverse modalities, plays a critical role in tumor biology and therapy response. Utilizing machine learning techniques, we developed a novel Programmed Cell Death Index (PCDI) incorporating multiple forms of PCD-related genes to predict outcomes in colorectal cancer CRC patients. The PCDI demonstrated robust prognostic performance, stratifying patients into high- and low-risk groups across multiple cohorts, with high PCDI scores correlating with poor survival, advanced tumor stage, and aggressive pathological features. A nomogram integrating PCDI with clinical variables showed strong predictive accuracy for 1-, 3-, and 5 year survival rates. Functional analysis revealed significant metabolic differences between high- and low-PCDI groups. Immune profiling identified associations between PCDI and immunosuppressive microenvironments, including elevated regulatory T cell levels and reduced PD-L1 expression in high-PCDI patients. Patients with high PCDI exhibited a potential resistance to immune checkpoint inhibitors. These findings emphasize PCDI's potential as a prognostic biomarker and a tool for guiding personalized therapeutic strategies in CRC patients.
Collapse
Affiliation(s)
- Jinping Li
- Yangzhou Polytechnic College, Yangzhou, 225009, Jiangsu, China
| | - Yan Jiang
- Department of Nephrology, Youjiang Medical College for Nationalities Affiliated Hospital, Baise, 533000, China
- Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi (Cultivation), Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Shengbin Nong
- Department of Nephrology, Youjiang Medical College for Nationalities Affiliated Hospital, Baise, 533000, China
- Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi (Cultivation), Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Liudan Liang
- Department of Nephrology, Youjiang Medical College for Nationalities Affiliated Hospital, Baise, 533000, China
- Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi (Cultivation), Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Liangchao Chen
- Department of Oncology, Xichong People's Hospital, Nanchong, 637200, China.
| | - Qiming Gong
- Department of Nephrology, Youjiang Medical College for Nationalities Affiliated Hospital, Baise, 533000, China.
- Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi (Cultivation), Youjiang Medical University for Nationalities, Baise, 533000, China.
| |
Collapse
|
29
|
Gupta G, Samuel VP, M RM, Rani B, Sasikumar Y, Nayak PP, Sudan P, Goyal K, Oliver BG, Chakraborty A, Dua K. Caspase-independent cell death in lung cancer: from mechanisms to clinical applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04149-0. [PMID: 40257494 DOI: 10.1007/s00210-025-04149-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/05/2025] [Indexed: 04/22/2025]
Abstract
Caspase-independent cell death (CICD) has recently become a very important mechanism in lung cancer, in particular, to overcome a critical failure in apoptotic cell death that is common to disease progression and treatment failures. The pathways involved in CICD span from necroptosis, ferroptosis, mitochondrial dysfunction, and autophagy-mediated cell death. Its potential therapeutic applications have been recently highlighted. Glutathione peroxidase 4 (GPX4) inhibition-driven ferroptosis has overcome drug resistance in non-small cell lung cancer (NSCLC). In addition, necroptosis involving RIPK1 and RIPK3 causes tumor cell death and modulation of immune responses in the tumor microenvironment (TME). Mitochondrial pathways are critical for CICD through modulation of metabolic and redox homeostasis. Ferroptosis is amplified by mitochondrial reactive oxygen species (ROS) and lipid peroxidation in lung cancer cells, and mitochondrial depolarization induces oxidative stress and leads to cell death. In addition, mitochondria-mediated autophagy, or mitophagy, results in the clearance of damaged organelles under stress conditions, while this function is also linked to CICD when dysregulated. The role of cell death through autophagy regulated by ATG proteins and PI3K/AKT/mTOR pathway is dual: to suppress tumor and to sensitize cells to therapy. A promising approach to enhancing therapeutic outcomes involves targeting mechanisms of CICD, including inducing ferroptosis by SLC7A11 inhibition, modulating mitochondrial ROS generation, or combining inhibition of autophagy with chemotherapy. Here, we review the molecular underpinnings of CICD, particularly on mitochondrial pathways and their potential to transform lung cancer treatment.
Collapse
Affiliation(s)
- Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vijaya Paul Samuel
- Department of Anatomy, RAK College of Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Bindu Rani
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Y Sasikumar
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Priya Priyadarshini Nayak
- Department of Medical Oncology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Puneet Sudan
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Brian G Oliver
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Amlan Chakraborty
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Kamal Dua
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia.
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
30
|
Taghizadieh M, Kalantari M, Bakhshali R, Kobravi S, Khalilollah S, Baghi HB, Bayat M, Nahand JS, Akhavan-Sigari R. To be or not to be: navigating the influence of MicroRNAs on cervical cancer cell death. Cancer Cell Int 2025; 25:153. [PMID: 40251577 PMCID: PMC12008905 DOI: 10.1186/s12935-025-03786-y] [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: 02/29/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
With all diagnostic and therapeutic advances, such as surgery, radiation- and chemo-therapy, cervical cancer (CC) is still ranked fourth among the most frequent cancers in women globally. New biomarkers and therapeutic targets are warranted to be discovered for the early detection, treatment, and prognosis of CC. As component of the non-coding RNA's family, microRNAs (miRNAs) participate in several cellular functions such as cell proliferation, gene expression, many signaling cascades, apoptosis, angiogenesis, etc. MiRNAs can suppress or induce programmed cell death (PCD) pathways by altering their regulatory genes. Besides, abnormal expression of miRNAs weakens or promotes various signaling pathways associated with PCD, resulting in the development of human diseases such as CC. For that reason, understanding the effects that miRNAs exert on the various modes of tumor PCD, and evaluating the potential of miRNAs to serve as targets for induction of cell death and reappearance of chemotherapy. The current study aims to define the effect that miRNAs exert on cell apoptosis, autophagy, pyroptosis, ferroptosis, and anoikis in cervical cancer to investigate possible targets for cervical cancer therapy. Manipulating the PCD pathways by miRNAs could be considered a primary therapeutic strategy for cervical cancer.
Collapse
Affiliation(s)
- Mohammad Taghizadieh
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Kalantari
- Department of Biology, Tehran University of health Sciences, Tehran, Iran
| | | | - Sepehr Kobravi
- Department of Oral & Maxillofacial Surgery, Faculty of Dentistry, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Shayan Khalilollah
- Department of Neurosurgery, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
31
|
Li H, Zhou D, Wang W, Aimaier R, JunYang, Zhao H, Zhao H, Li J, Pang X, Zhou Q, Zhao H. Mycosubtilin Induces G1 Phase Block and Autophagy in Cervical Cancer HeLa Cells. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10534-1. [PMID: 40240746 DOI: 10.1007/s12602-025-10534-1] [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] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Cyclic lipopeptides secreted by the probiotic bacterium Bacillus subtilis have attracted much attention due to their antitumor activities and low toxicity. However, the role of Mycosubtilin (Myco) in the prevention and treatment of cervical cancer remains unclear. In the present study, we conducted a systematic evaluation of Myco's anti-cervical cancer effects to identify its molecular mechanism of action using proteomics technology. The results reveal that Myco inhibited the growth of HeLa and SiHa cervical cancer cell lines in a dose-dependent (3-15 µg/mL) and time-dependent (12-48 h) manner and significantly reduced colony formation and migration in HeLa cells, highlighting its potential to suppress tumor spread. Moreover, autophagosome and autolysosome numbers were significantly increased after Myco treatment, and the expression of autophagy-related proteins was significantly modulated, suggesting that autophagy plays a role in its anti-cancer mechanism. Myco treatment also induced G1 phase cell cycle arrest in HeLa cells, as confirmed by proteomics analysis. Myco was shown to induce cell cycle arrest in HeLa cells by regulating the P53 pathway and autophagy-dependent cell death via the PI3K/AKT/mTOR signaling pathway, demonstrating its multidimensional effect on cervical cancer cell growths. Myco treatment significantly inhibited tumor growth in vivo in a nude mouse cervical cancer xenograft model, providing direct evidence of its potential as a therapeutic candidate for cervical cancer. Given its unique anti-cancer mechanism and significant therapeutic efficacy, Myco should be considered a promising therapeutic agent for cervical cancer.
Collapse
Affiliation(s)
- Haoran Li
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Dongyuan Zhou
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Weiquan Wang
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Reyihanguli Aimaier
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
| | - JunYang
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Haoran Zhao
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Heping Zhao
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jinyu Li
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Xiufeng Pang
- Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Qian Zhou
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China.
| | - Huixin Zhao
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China.
| |
Collapse
|
32
|
Jiang P, Jiang Z, Li S, Li YX, Chen Y, Li X. The suppressive role of GLS in radiosensitivity and irradiation-induced immune response in LUAD: integrating bioinformatics and experimental insights. Front Immunol 2025; 16:1582587. [PMID: 40308578 PMCID: PMC12040943 DOI: 10.3389/fimmu.2025.1582587] [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: 02/24/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Background Radiotherapy elicits immune activation, thereby synergistically enhancing systemic tumor control when combined with immunotherapy. Glutaminase (GLS), a key enzyme for glutamine metabolism, has been found to regulate glutamine availability within tumor microenvironment (TME). However, the precise mechanisms through which GLS modulates radiosensitivity and irradiation-induced immune responses in lung adenocarcinoma (LUAD) and its clinical value remain to be fully elucidated. Methods We employed bulk RNA-seq and single-cell transcriptomics to explore the role of GLS expression in radiosensitivity and immune infiltration. The bioinformatic results were validated by in vitro and in vivo experiments. Co-culture assays and flow cytometry were used to validate the impact of GLS expression on CD8+ T cell activation and cytotoxicity. Moreover, a GLS-DSBr (double strand break repair) prognostic model was developed using machine learning with data from 2,066 LUAD patients. Results In vitro and in vivo experiments demonstrated that GLS silence inhibited DSB repair and promoted ferroptosis, therefore enhancing radiosensitivity. Single-cell and spatial transcriptomics revealed the immunomodulatory effects of GLS expression in the TME. Further, Co-culture assays and flow cytometry experiments indicated that silencing GLS in LUAD cells potentiated the activation and cytotoxicity of CD8+ T cells in the context of radiotherapy. The GLS-DSBr model demonstrated robust predictive performance for overall survival, as well as the efficacy of radiotherapy and immunotherapy in LUAD. The applicability of GLS-DSBr model was further validated through pan-cancer analysis. Conclusion In the contexts of radiotherapy, GLS downregulation exerts dual regulatory effects by modulating ferroptosis and remodeling the immune landscapes, particularly enhancing CD8+ T cell cytotoxicity. Our work suggests that strategies preferentially targeting GLS in tumor cells may represent promising and translatable therapeutic approaches to promote antitumor efficacy of radiotherapy plus immune checkpoint blockade in LUAD patients. Furthermore, the established GLS-DSBr model serves as a robust predictive tool for prognosis and effects of radiotherapy and immunotherapy, which assists personalized treatment optimization in LUAD.
Collapse
Affiliation(s)
- Peicheng Jiang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhifeng Jiang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Su Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Department of Cardiology, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Ye-Xiong Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuqiong Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Xinyan Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
Chen L, Liu Y, Li H, Lin S, Wang X, Fang J, Diao X, Wang L, Yang Z, Cai Z. Size-Dependent Pulmonary Toxicity and Whole-Body Distribution of Inhaled Micro/Nanoplastic Particles in Male Mice from Chronic Exposure. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:6993-7003. [PMID: 40181497 DOI: 10.1021/acs.est.4c14232] [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
The ubiquitous presence of micro/nanoplastics (MP/NP) in the atmosphere has raised significant concerns about their potential health risks through inhalation, yet the effects of natural respiratory exposure remain underexplored. This study addresses this critical knowledge void by utilizing a whole-body inhalation exposure system to investigate the distribution, accumulation, and pulmonary toxicity of polystyrene MP/NP (1.5 × 105 particles/m3) in male ICR mice (n = 16/group). Fluorescently labeled MP/NP revealed the highest particle accumulation in the lungs, followed by the bloodstream and spleen, with minimal detection in the brain. Unsurprisingly, 80 nm nanoplastics displayed greater intertissue transport efficiency than 1 μm microplastics. Chronic exposure to both microplastics and nanoplastics disrupted oxidative balance and exacerbated oxidative stress within the extracellular environment of the lungs. The impaired antioxidant defenses and disrupted intra- and extracellular metabolism led to inflammation, apoptosis, and fibrosis. Intriguingly, 1 μm microplastics induced more severe pulmonary toxicity than their smaller counterparts, promoting epithelial-mesenchymal transition and fibrosis. These findings underscore the need for a nuanced understanding of size-dependent toxicities of inhalable plastic particles and highlight the health risks posed by airborne MP/NP.
Collapse
Affiliation(s)
- Leijian Chen
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Yu Liu
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Huankai Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Siyi Lin
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Xiaoxiao Wang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Jiacheng Fang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Xin Diao
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Lei Wang
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Zhu Yang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
- Department of Biology, Hong Kong Baptist University, Hong Kong 999077, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| |
Collapse
|
34
|
Xiang Z, Mei H, Wang H, Yao X, Rao J, Zhang W, Xu A, Lu L. Cuproptosis and its potential role in musculoskeletal disease. Front Cell Dev Biol 2025; 13:1570131. [PMID: 40292330 PMCID: PMC12022686 DOI: 10.3389/fcell.2025.1570131] [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: 02/03/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Cuproptosis, a recently identified form of copper-dependent cell death, arises from intracellular copper dyshomeostasis. As an essential trace element, copper plays a critical role in bioenergetic metabolism, redox regulation, and synaptic transmission. However, excessive copper exerts cytotoxic effects through multiple pathways, including increased reactive oxygen species (ROS) production, apoptotic cascade activation, necrotic membrane rupture, inflammatory responses, and mitochondrial dysfunction. Distinct from other cell death mechanisms, cuproptosis is characterized by copper ion binding to acetylated mitochondrial respiratory chain proteins, leading to pathogenic protein aggregation, iron-sulfur cluster depletion, and cellular collapse. Emerging evidence underscores aberrant copper accumulation and resultant proteotoxic stress as pivotal contributors to the pathogenesis of multiple musculoskeletal pathologies, including osteoporosis, osteoarthritis, sarcopenia, osteosarcoma, intervertebral disc degeneration, spinal cord injury, and biofilm-associated orthopedic infections. Understanding the spatiotemporal regulation of cuproptosis may provide novel opportunities for advancing diagnostic and therapeutic approaches in orthopedic medicine. This review synthesizes current insights into the molecular mechanisms of cuproptosis, its pathogenic role in musculoskeletal diseases, and the potential for biomarker-driven therapeutic interventions.
Collapse
Affiliation(s)
- Ziyang Xiang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huiling Mei
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Honglin Wang
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyue Yao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ji Rao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wentao Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Aoshuang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
35
|
Muszka Z, Jenei V, Mácsik R, Mezhonova E, Diyab S, Csősz R, Bácsi A, Mázló A, Koncz G. Life-threatening risk factors contribute to the development of diseases with the highest mortality through the induction of regulated necrotic cell death. Cell Death Dis 2025; 16:273. [PMID: 40216765 PMCID: PMC11992264 DOI: 10.1038/s41419-025-07563-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025]
Abstract
Chronic diseases affecting the cardiovascular system, diabetes mellitus, neurodegenerative diseases, and various other organ-specific conditions, involve different underlying pathological processes. However, they share common risk factors that contribute to the development and progression of these diseases, including air pollution, hypertension, obesity, high cholesterol levels, smoking and alcoholism. In this review, we aim to explore the connection between four types of diseases with different etiologies and various risk factors. We highlight that the presence of risk factors induces regulated necrotic cell death, leading to the release of damage-associated molecular patterns (DAMPs), ultimately resulting in sterile inflammation. Therefore, DAMP-mediated inflammation may be the link explaining how risk factors can lead to the development and maintenance of chronic diseases. To explore these processes, we summarize the main cell death pathways activated by the most common life-threatening risk factors, the types of released DAMPs and how these events are associated with the pathophysiology of diseases with the highest mortality. Various risk factors, such as smoking, air pollution, alcoholism, hypertension, obesity, and high cholesterol levels induce regulated necrosis. Subsequently, the release of DAMPs leads to chronic inflammation, which increases the risk of many diseases, including those with the highest mortality rates.
Collapse
Affiliation(s)
- Zsuzsa Muszka
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Viktória Jenei
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Rebeka Mácsik
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Evgeniya Mezhonova
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Silina Diyab
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Réka Csősz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| |
Collapse
|
36
|
Zhang Z, Rana I, Nam J. Metal coordination polymer nanoparticles for cancer therapy. Essays Biochem 2025; 69:EBC20253012. [PMID: 40209056 DOI: 10.1042/ebc20253012] [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: 02/25/2025] [Accepted: 03/24/2025] [Indexed: 04/12/2025]
Abstract
Metal ions are essential elements in biological processes and immune homeostasis. They can regulate cancer cell death through multiple distinct molecular pathways and stimulate immune cells implicated in antitumor immune responses, suggesting opportunities to design novel metal ion-based cancer therapies. However, their small size and high charge density result in poor target cell uptake, uncontrolled biodistribution, and rapid clearance from the body, reducing therapeutic efficacy and increasing potential off-target toxicity. Metal coordination polymer nanoparticles (MCP NPs) are nanoscale polymer networks composed of metal ions and organic ligands linked via noncovalent coordination interactions. MCP NPs offer a promising nanoplatform for reshaping metal ions into more drug-like formulations, improving their in vivo pharmacological performance and therapeutic index for cancer therapy applications. This review provides a comprehensive overview of the inherent biological functions of metal ions in cancer therapy, showcasing examples of MCP NP systems designed for preclinical cancer therapy applications where drug delivery principles play a critical role in enhancing therapeutic outcomes. MCP NPs offer versatile metal ion engineering approaches using selected metal ions, various organic ligands, and functional payloads, enabling on-demand nano-drug designs that can significantly improve therapeutic efficacy and reduce side effects for effective cancer therapy.
Collapse
Affiliation(s)
- Zhengzheng Zhang
- College of Pharmacy, Chonnam National University, Gwanju 61186, South Korea
| | - Isra Rana
- College of Pharmacy, Chonnam National University, Gwanju 61186, South Korea
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwanju 61186, South Korea
| |
Collapse
|
37
|
Mohamed HRH, Essam R, Mohamed BA, Hakeem GM, Elnawasani SH, Nagy M, Safwat G, Diab A. Potent cytotoxicity and induction of ROS-mediated genomic instability, mitochondrial dysfunction, and apoptosis by Y 2O 3 NPs in Hep-G2 hepatic cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04051-9. [PMID: 40208319 DOI: 10.1007/s00210-025-04051-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025]
Abstract
Hepatic cancer, one of the most prevalent and lethal cancers globally, remains a significant health challenge, with limited treatment options underscoring the urgent need for novel, more effective therapies. Yttrium oxide nanoparticles (Y2O3 NPs) have attracted attention in nanomedicine due to their promising properties, including enhanced drug delivery, imaging capabilities, and therapeutic effects. However, the specific impact of Y2O3 NPs on hepatic cancer is largely unexplored. Therefore, this study was conducted to assess the cytotoxic effects of Y2O3 NPs on cell viability, reactive oxygen species (ROS) generation, genomic stability, mitochondrial integrity, and apoptosis induction in Hep-G2 hepatic cancer cells. The results from the SRB cytotoxicity assay demonstrated a strong concentration-dependent decrease in Hep-G2 cell viability, with a notably low half-maximal inhibitory concentration (IC50) value of 13.15 µg/ml. Exposure to the IC50 concentration of Y2O3 NPs led to increased ROS generation, DNA damage induction, and loss of mitochondrial membrane potential. Furthermore, the expression of pro-apoptotic p53 and mitochondrial ND3 genes was significantly upregulated, while the anti-apoptotic Bcl-2 gene was markedly downregulated, triggering apoptosis in Hep-G2 cells after 72 h of exposure to Y2O3 NPs. Collectively, these findings highlight the therapeutic potential of Y2O3 NPs in hepatic cancer, emphasizing the need for further research to fully explore their efficacy as a treatment option for liver cancer.
Collapse
Affiliation(s)
- Hanan R H Mohamed
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Rawan Essam
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6Th of October City, Egypt
| | - Basma A Mohamed
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6Th of October City, Egypt
| | - George M Hakeem
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6Th of October City, Egypt
| | - Shahd H Elnawasani
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6Th of October City, Egypt
| | - Maria Nagy
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6Th of October City, Egypt
| | - Gehan Safwat
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6Th of October City, Egypt
| | - Ayman Diab
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6Th of October City, Egypt
| |
Collapse
|
38
|
Peng Y, Jia N, Wang J, Dong S, Li S, Qin W, Shi H, Liu K. Analysis of Multiple Programmed Cell Death Patterns and Functional Validations of Apoptosis-Associated Genes in Lung Adenocarcinoma. Ann Surg Oncol 2025:10.1245/s10434-025-17224-w. [PMID: 40175857 DOI: 10.1245/s10434-025-17224-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/09/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is marked by its considerable aggressiveness and pronounced heterogeneity. Programmed cell death (PCD) plays a pivotal role in the progression of tumors, their aggressive behavior, resistance to treatment, and recurrence of the disease. PATIENTS AND METHODS Using expression data from 878 patients across four multicenter cohorts, we identified 13 consensus prognostic genes from 1481 genes associated with PCD. We employed 10 machine-learning algorithms, generating 101 combinations, from which the optimal algorithm was chosen to develop an artificial intelligence-derived cell death index (CDI) on the basis of the average C-index. RESULTS The training cohort and three external validation cohorts consistently demonstrated that CDI could accurately predict LUAD prognosis. Moreover, CDI showed significantly greater accuracy than traditional clinical variables, molecular characteristics, and 22 previously published signatures. Patients in the low-CDI group had a more favorable prognosis, higher levels of immune cell infiltration, better responsiveness to immunotherapy, and a higher likelihood of displaying the "hot tumor" phenotype. Single-cell analysis revealed that neutrophils had the highest CDI scores and exhibited significant differences in marker gene expression. CONCLUSIONS Pseudotime trajectory analysis indicated that BCL2L14 plays a crucial role in the developmental pathway of neutrophils, potentially influencing the fate of LUAD cells. Knockdown of BCL2L14 significantly reduced the growth, proliferation, and colony formation abilities of LUAD cells, while also enhancing apoptosis rates.
Collapse
Affiliation(s)
- Yu Peng
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Nan Jia
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, China
| | - Jingyu Wang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Shilei Dong
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Shujun Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Wei Qin
- Internal Medicine of Integrated Chinese and Western Medicine, Affiliated Hospital of Hebei University, Baoding, China.
| | - Hongyun Shi
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China.
| | - Kuan Liu
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China.
| |
Collapse
|
39
|
R RJ, Choudhury B, Alam MM, Mm B, Chanda K. Unlocking the power of imidazoquinolines: recent advances in anticancer and immunotherapeutic strategies. Future Med Chem 2025; 17:943-959. [PMID: 40231819 DOI: 10.1080/17568919.2025.2491303] [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: 01/13/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
The challenges in drug discovery aiming to mitigate cancer progression are the thrust area of scientific research for several decades. Since the advent of heterocyclic chemistry, drug discovery programs have made significant achievements that lead to the development of numerous drugs with broad spectrum of potencies, contributing to both diagnostic and therapeutic advancements. Till date, efforts to discover more potent and efficient drug candidates are underway to minimize adverse side effects of existing chemotherapeutics. In view of the above, small-molecule agonists that can interact with different immune modulators like toll like receptor-7 (TLR-7) and TLR-8 are being investigated and explored. These candidates are expected to display profound effect on anti-tumoral activity by enhancing the production of proinflammatory cytokines. Recently, numerous imidazoquinoline derivatives with proven TLR agonist activities have emerged as promising anticancer therapeutics. With advancements in technology and the evolution of new scopes in drug discovery, different strategies are being adopted, particularly with the help of nanotechnology, immune-technology, combination drug chemistry, etc., to curb the progression of various types of cancers. Herein, the novel strategies for cancer therapeutics with imidazoquinolines reported in the last 5 years, their structure-activity relationship along with important synthetic schemes for important TLR agonists, are discussed.
Collapse
Affiliation(s)
- Ranjini Jenifer R
- Chemistry Division, School of Advanced Sciences, Vellore Institute of Technology, Chennai, India
| | - Badruzzaman Choudhury
- Department of Chemistry, School of Advanced Sciences, Vellore institute of Technology, Vellore, India
| | - Mohammed Mujahid Alam
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Balamurali Mm
- Chemistry Division, School of Advanced Sciences, Vellore Institute of Technology, Chennai, India
| | - Kaushik Chanda
- Department of Chemistry, Rabindranath Tagore University, Hojai, India
| |
Collapse
|
40
|
Zhang W, Shan G, Bi G, Hu Z, Yi Y, Zeng D, Lin Z, Zhan C. Lactylation and regulated cell death. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119927. [PMID: 40023198 DOI: 10.1016/j.bbamcr.2025.119927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Lactylation, a newly identified post-translational modification, entails the attachment of lactate to lysine residues within proteins, profoundly modulating diverse cellular mechanisms underlying regulated cell death (RCD). This modification encompasses two primary categories: histone lactylation and non-histone lactylation. Histone lactylation assumes a pivotal regulatory function in the RCD process, primarily by modulating the transcriptional landscape of genes implicated in cell death. In contrast, non-histone lactylation exerts its influence by targeting transferases, transcription, cell cycle progression, death pathways, and metabolic processes that are intricately involved in RCD. This review provides a comprehensive overview of recent breakthroughs in understanding how lactylation regulates RCD, while also offering insights into potential avenues for future research, thereby deepening our comprehension of cellular fate determination.
Collapse
Affiliation(s)
- Wenlong Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Yanjun Yi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Dejun Zeng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Zongwu Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China.
| |
Collapse
|
41
|
Yang MH, Son NT, Bastos JK, Luyen ND, Linh NN, Ahn KS. Eriodictyol 5-O-methyl ether inhibits prostate cancer progression through targeting STAT3 signaling and inducing apoptosis and paraptosis. Arch Biochem Biophys 2025; 766:110331. [PMID: 39924141 DOI: 10.1016/j.abb.2025.110331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/20/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Prostate cancer ranks as one of the most prevalent cancers among men and is a major cause of cancer-related mortality globally This study aims to elucidate the molecular mechanisms underlying the anti-cancer effects of eriodictyol 5-O-methyl ether (ERIO) on prostate cancer cells, focusing on its impact on STAT3 signaling, apoptosis, and paraptosis. ERIO exhibited significant cytotoxicity against DU145, PC-3, and LNCaP cells. It suppressed constitutive and IL-6-induced STAT3 activation by inhibiting the phosphorylation of JAK1, JAK2, and Src kinases. ERIO upregulated SHP-2 expression, leading to the dephosphorylation of STAT3. ERIO induced apoptosis, evidenced by increased caspase-3 and PARP cleavage, and paraptosis, characterized by increased ROS production, decreased mitochondrial membrane potential, and ER stress. The antioxidant NAC reversed the effects of ERIO, highlighting the importance of oxidative stress in its anti-cancer activity. ERIO effectively inhibited prostate cancer cell growth by targeting STAT3 signaling and inducing both apoptosis and paraptosis. These findings suggest that ERIO has significant therapeutic potential for prostate cancer treatment and warrant further investigation in in vivo and clinical studies.
Collapse
Affiliation(s)
- Min Hee Yang
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Ninh The Son
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), Hoang Quoc Viet, Caugiay, Hanoi, 10000, Viet Nam; Department of Chemistry, Graduate University of Science and Technology, VAST, Hoang Quoc Viet, Caugiay, Hanoi, 10000, Viet Nam; University of São Paulo (USP), School of Pharmaceutical Sciences of Ribeirão Preto, SP, Brazil.
| | - Jairo Kenupp Bastos
- University of São Paulo (USP), School of Pharmaceutical Sciences of Ribeirão Preto, SP, Brazil.
| | - Nguyen Dinh Luyen
- Institute of Natural Products Chemistry, VAST, Hoang Quoc Viet, Caugiay, Hanoi, 10000, Viet Nam.
| | - Nguyen Ngoc Linh
- Faculty of Pharmacy, Thanh Do University, Kim Chung, Hoai Duc, Hanoi, 10000, Viet Nam.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
42
|
Karankar VS, Awasthi S, Srivastava N. Peptide-driven strategies against lung cancer. Life Sci 2025; 366-367:123453. [PMID: 39923837 DOI: 10.1016/j.lfs.2025.123453] [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: 11/11/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Lung cancer remains one of the most significant global health challenges, accounting for 18 % of all cancer-related deaths. While risk factors such as heavy metal exposure and cigarette smoking are well-known contributors, the limitations of conventional treatments including severe side effects and drug resistance highlight the urgent need for more targeted and safer therapeutic options. In this context, peptides have emerged as a novel, precise, and effective class of therapies for lung cancer treatment. They have shown promise in limiting lung cancer progression by targeting key molecular pathways involved in tumour growth. Anti-non-small cell lung cancer peptides that specifically target proteins such as EGFR, TP53, BRAF, MET, ROS1, and ALK have demonstrated potential in improving lung cancer outcomes. Additionally, anti-inflammatory and apoptosis-inducing peptides offer further therapeutic benefits. This review provides a comprehensive overview of the peptides currently in use or under investigation for the treatment of lung cancer, highlighting their mechanisms of action and therapeutic potential. As research continues to advance, peptides are poised to become a promising new therapeutic option in the fight against lung cancer.
Collapse
Affiliation(s)
- Vijayshree S Karankar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Saurabh Awasthi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India.
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India.
| |
Collapse
|
43
|
Cheng L, Wang Y, Zhang Y. Dying to survive: harnessing inflammatory cell death for better immunotherapy. Trends Cancer 2025; 11:376-402. [PMID: 39986988 DOI: 10.1016/j.trecan.2025.01.012] [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: 10/01/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/24/2025]
Abstract
Immunotherapy has transformed cancer treatment paradigms, but its effectiveness depends largely on the immunogenicity of the tumor. Unfortunately, the high resemblance of cancer to normal tissues makes most tumors immunologically 'cold', with a poor response to immunotherapy. Danger signals are critical for breaking immune tolerance and mobilizing robust, long-lasting antitumor immunity. Recent studies have identified inflammatory cell death modalities and their power in providing danger signals to trigger optimal tumor suppression. However, key mediators of inflammatory cell death are preferentially silenced during early tumor immunoediting. Strategies to rejuvenate inflammatory cell death hold great promise for broadening immunotherapy-responsive tumors. In this review, we examine how inflammatory cell death enhances tumor immunogenicity, how it is suppressed during immunoediting, and the potential of harnessing it for improved immunotherapy.
Collapse
Affiliation(s)
- Long Cheng
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Yibo Wang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ying Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
44
|
Liu J, Qiu XR, Tian YL, Sun WJ, Wang YH, Liu H, Zhang G, Zhao WZ, Yin DC, Zhang CY. Urchin-like magnetic nanoparticles loaded with type X collagen siRNA and Stattic to treat triple negative breast cancer under rotating magnetic field like an "enchanted micro-scalpel". Int J Biol Macromol 2025; 300:140318. [PMID: 39864683 DOI: 10.1016/j.ijbiomac.2025.140318] [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: 10/27/2024] [Revised: 01/11/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
Magnetic nanoparticles effectively target drug delivery, contrast agents, biosensors, and more. Urchin-like magnetic nanoparticles (UMN) with abundant spike-like structures exhibit superior magneto-mechanical force to destroy tumor cells compared with other shapes of magnetic nanoparticles. However, when cell contents are released from tumor cells induced by magneto-mechanical force, they can act on surrounding tumor cells to facilitate tumor development. Therefore, multifunctional UMN is necessary. Interleukin-6 (IL-6) is an important inflammatory factor which is released after cell rupture, it can activate the STAT3 signaling pathway to promote tumor progression. Type X collagen (COL10A1) is a significant component of the extracellular matrix, ranking second among all aberrant genes in triple negative breast cancer (TNBC), and its knockdown can suppress tumorigenesis and metastasis. Here, we built a rotating magnetic field (RMF) platform, and a novel UMN using a straightforward solvothermal method was synthesized, which was much simpler than existing method. Stattic (STAT3 inhibitor) and COL10A1 siRNA were loaded onto the UMN@PEI to form UMNP/St/si. The RMF drove UMNP/St/si disrupted the cell membrane, promoted cell death. The inhibitory effects of UMNP/St/si under RMF on TNBC were verified both in vitro and in vivo. Furthermore, despite the increase in IL-6 due to cell rupture, IL-6/STAT3 signaling pathway was inhibited by Stattic, compensating for the deficiency of magneto-mechanical force. Moreover, the underlying mechanical mechanism of UMNP/St/si after exposure to RMF was also analyzed. It suggests that UMNP/St/si is a promising and effective strategy for TNBC treatment and provides valuable insights for treating other diseases as well.
Collapse
Affiliation(s)
- Jie Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China
| | - Xiao-Rong Qiu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China
| | - Yi-Le Tian
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China
| | - Wen-Jun Sun
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China
| | - Yu-Hua Wang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China
| | - Huan Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China
| | - Wen-Zhe Zhao
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710100, Shaanxi, PR China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518063, PR China.
| |
Collapse
|
45
|
Shangguan F, Ma N, Chen Y, Zheng Y, Ma T, An J, Lin J, Yang H. Fucoxanthin suppresses pancreatic cancer progression by inducing bioenergetics metabolism crisis and promoting SLC31A1‑mediated sensitivity to DDP. Int J Oncol 2025; 66:31. [PMID: 40052552 PMCID: PMC11900939 DOI: 10.3892/ijo.2025.5737] [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: 09/06/2024] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
Pancreatic cancer (PC) is one of the most malignant tumors, with a 5‑year survival rate <10%. Chemosynthetic drugs are widely used in the treatment of PC; however, their toxicity and side effects often reduce the quality of life for patients. MTT and colony formation assay were performed to detect cell growth and viability in PC cells. Levels of ROS in whole cell and mitochondria were analyzed through flow cytometry. ATP production was evaluated using an ATP Assay Kit. Cellular bioenergetics were analyzed with a Seahorse XFe96 Analyzer, and changes in target molecules were monitored by western blotting. The present study reports that fucoxanthin (FX), a carotenoid derived from aquatic brown seaweed, significantly inhibits PC by inhibiting cell proliferation and inducing cell death via the non‑classical pathway. FX switches mitochondrial respiration to aerobic glycolysis in PC cells. Furthermore, FX decreases whole‑cell ATP levels, which indicates that promotion of glycolysis does not compensate for FX‑induced ATP depletion in mitochondria. Moreover, FX decreased the reduced glutathione/oxidized glutathione ratio observed under glucose‑limited conditions. These alterations caused by FX may decrease metabolic flexibility, indicating higher sensitivity to glucose‑limited (GL) conditions. FX increased the cytotoxicity of cisplatin (DDP) and the expression of solute carrier family 31 member 1 (SLC31A1) in PC cells. Furthermore, the knockdown of SLC31A1 can attenuate cytotoxicity caused by the combination of FX and DDP. It was inferred that FX increased the sensitivity of PC cells to DDP), potentially by upregulating SLC31A1 expression. In conclusion, FX exhibited potent antitumor effects by reprogramming energy metabolism and inducing a distinct form of regulated cell death. Therefore, combining FX with GL treatment or DDP presents a promising therapeutic strategy for PC.
Collapse
Affiliation(s)
- Fugen Shangguan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Nengfang Ma
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yang Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuansi Zheng
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Ting Ma
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jing An
- Division of Infectious Diseases and Global Health, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Jianhu Lin
- Department of Trauma Surgery and Emergency Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Hailong Yang
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
46
|
Akhavan R, Jabari Harsini S, Shafiee S, Eftekhari M, Tahmasvand R, Taghipour F, Kooshafar Z, Mohammadmirzaeizarandi H, Salimi M, Almasirad A, Salimi M. Discovery of a new hydrazone-oxamide hybrid capable of inducing necroptotic cell death in triple negative breast cancer cells. Bioorg Chem 2025; 157:108267. [PMID: 39986105 DOI: 10.1016/j.bioorg.2025.108267] [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: 10/07/2024] [Revised: 01/22/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
The poor prognosis and inefficiency of the therapeutic agents in treating triple negative breast cancer (TNBC) have raised significant concerns, driving the quest for designing novel and potent chemotherapeutic compounds. In this regard, inducing programmed cell death (PCD) has emerged as a promising approach for breast cancer therapy. Accordingly, a series of hybrid molecules comprising hydrazone and oxamide moieties (5a-5q) were designed, synthesized, and assessed for their anticancer activity against various cancer cells. Among these synthesized hybrids, compound 5q was selected as the lead compound with remarkable ability to disrupt MDA-MB-231 cell growth, achieving an IC50-72h of 9.79 μM, while exhibiting lower toxicity in normal human cells. The in vitro experiments revealed that this compound triggers neither apoptosis nor autophagy in TNBC cells. Furthermore, the in vivo outcomes corroborated the in vitro results, showing a significant delay in tumor growth at a dose of 1 mg/kg/day following three weeks of treatment in the 4T1 mouse model of TNBC. The findings of this study suggested that compound 5q acts through necroptosis by overexpression of P-RIPK3 and phosphorylation of its downstream effector, MLKL. Compound 5q holds promise as a potential candidate for the development of anti-TNBC drugs.
Collapse
Affiliation(s)
- Romina Akhavan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Sanaz Jabari Harsini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Samira Shafiee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahya Eftekhari
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Raheleh Tahmasvand
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Fereshteh Taghipour
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Kooshafar
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Hiva Mohammadmirzaeizarandi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Misha Salimi
- Department of Biology, Faculty of Converging Sciences and Technology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ali Almasirad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mona Salimi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
47
|
Chota A, Abrahamse H, George BP. Chemotoxic and phototoxic effects of liposomal co-delivery of green synthesized silver nanoparticles and ZnPcS 4 for enhanced photodynamic therapy in MCF-7 breast cancer cells: An in vitro study. Biomed Pharmacother 2025; 185:117986. [PMID: 40090284 DOI: 10.1016/j.biopha.2025.117986] [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: 02/14/2025] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025] Open
Abstract
Breast cancer remains a significant challenge in oncology, despite notable advances in treatment methods. Traditional therapies such as surgery, chemotherapy, radiation, and hormonal treatments have long been used to manage breast cancer. However, often patients experience treatment failure, resulting in disease recurrence and progression. Therefore, this study explores the therapeutic potential of green-synthesized silver nanoparticles (AgNPs), using the root methanol (MeOH) extract of the African medicinal plant Dicoma anomala (D. anomala) as a reducing agent, to combat breast cancer. AgNPs were synthesized using a bottom-up approach and later modified with liposomes (Lip) loaded with the photosensitizer zinc phthalocyanine tetrasulfonate (Lip@ZnPcS4) through the thin film hydration method. Prior to in vitro cell culture studies, UV-Vis spectroscopy was used to study the in vitro drug release kinetics of nanoparticles (NPs) at pH 5.8 and 7.4 respectively. After a 24 h treatment period, MCF-7 breast cancer cells were evaluated for cell cytotoxicity using lactate dehydrogenase Cyto-Tox96® Non-Radioactive Cytotoxicity Assay Kit LDH and cell viability using the CellTiter-Glo® ATP luminescence assay kit. Cell death studies were analyzed using an inverted light microscope for morphological changes, fluorescence microscopy for reactive oxygen species (ROS) detection and Live/Dead cell viability, human p53 protein analysis using enzyme-linked immunosorbent assay (ELISA), apoptotic and anti-apoptotic protein analysis by immunofluorescence, and gene expression analysis using real-time reverse transcription polymerase chain reaction (RT-PCR) assay. The experiments were conducted in quadruplicate (n = 4), and the results were analyzed using IBM SPSS statistical software version 27, with a 95 % confidence interval. The synthesized NPs and nanocomplexes, including AgNPs, AgNPs-Lip, Lip@ZnPcS4, and AgNPs-Lip@ZnPcS4, demonstrated significant cytotoxicity and therapeutic potential against MCF-7 breast cancer cells. Notably, apoptosis was induced, primarily through the activation of the intrinsic pathway. Given the difficult prognosis associated with breast cancer, these findings highlight the promise of liposomal nanoformulations (NFs) in cancer photodynamic therapy (PDT), supporting further investigation in in vivo settings.
Collapse
Affiliation(s)
- Alexander Chota
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Blassan P George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa.
| |
Collapse
|
48
|
Xia X, Kong C, Zhao X, Zhao K, Shi N, Jiang J, Li P. The complexities of cell death mechanisms: a new perspective in systemic sclerosis therapy. Apoptosis 2025; 30:636-651. [PMID: 39924583 DOI: 10.1007/s10495-025-02082-4] [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] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
Systemic sclerosis, also termed scleroderma, is a severe and debilitating autoimmune disease characterized by fibrosis, an aberrant immune response, and vascular dysfunction. Cell death is essential to the body's continued normal development as it removes old or damaged cells. This process is governed by several mechanisms, including programmed cell death through apoptosis, necrosis, and pyroptosis, as well as metabolic processes, such as ferroptosis and cuproptosis. This review describes the signaling pathways associated with each form of cell death, examining the linkages between these pathways, and discussing how the dysregulation of cell death processes is involved in the development of autoimmune disorders such as systemic sclerosis. Existing and promising therapeutic strategies aimed at restoring the balance of cell death in systemic sclerosis and other autoimmune disorders are also emphasized.
Collapse
Affiliation(s)
- Xue Xia
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Chenfei Kong
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Xiaoming Zhao
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Kelin Zhao
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Naixu Shi
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, 130033, China.
| | - Ping Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China.
| |
Collapse
|
49
|
Wang Y, Sun X, Ren M, Ma F, Zhao R, Zhu X, Xu Y, Cao N, Chen Y, Pan Y, Zhao A. Integrative network pharmacology, transcriptomics, and proteomics reveal the material basis and mechanism of the Shen Qing Weichang Formula against gastric cancer. Chin Med 2025; 20:42. [PMID: 40155922 PMCID: PMC11954191 DOI: 10.1186/s13020-025-01091-4] [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: 11/19/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a common malignancy with poor prognosis and lack of efficient therapeutic methods. Shen Qing Weichang Formula (SQWCF) is a patented traditional herbal prescription for GC, but its efficacy and underlying mechanism remains to be clarified. PURPOSE To explore the efficacy and potential mechanism of SQWCF in treating GC. METHODS A subcutaneous transplantation tumor model of human GC was established for assessing SQWCF's efficacy and safety. A comprehensive strategy integrating mass spectrometry, network pharmacology, omics analysis, and bioinformatic methods was adopted to explore the core components, key targets, and potential mechanism of SQWCF in treating GC. Molecular docking, immunohistochemistry, quantitative real-time PCR, and western blot were applied to validation. RESULTS In the mouse model of GC, SQWCF effectively suppressed the GC growth without evident toxicity and enhanced the therapeutic efficacy of paclitaxel. Network pharmacology and molecular docking based on mass spectrometry showed that key targets (CASP3, TP53, Bcl-2, and AKT1) and core active components (Calycosin, Glycitein, Liquiritigenin, Hesperetin, and Eriodictyol) involved in the anti-GC effect of SQWCF had stable binding affinity, of which AKT1 ranked the top in the affinity. Validation based on network pharmacology and omics analysis confirmed that PI3K-AKT and MAPK signaling pathways, as well as downstream apoptosis pathway, explained the therapeutic effects of SQWCF on GC. In addition, family with sequence similarity 81 member A (FAM81A) was identified as a novel biomarker of GC that was aberrantly highly expressed in GC and associated with poor prognosis by bioinformatic analysis, and was an effector target of SQWCF at both mRNA and protein levels. CONCLUSION This study uncovers a synergistic multi-component, multi-target, and multi-pathway regulatory mechanism of SQWCF in treating GC comprehensively, emphasizing its potential for therapeutic use and providing new insights into GC treatment.
Collapse
Affiliation(s)
- Yi Wang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Xiaoyu Sun
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Mingming Ren
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Fangqi Ma
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Ruohan Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Xiaohong Zhu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Yan Xu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Nida Cao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Yuanyuan Chen
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China
| | - Yongfu Pan
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China.
| | - Aiguang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, South Wanping Rd. 725, Shanghai, 200032, China.
| |
Collapse
|
50
|
Ding M, Huo K, Chen X, Wang W, Xiang Z, Song Y, Chen P, Liu L. The role of non-coding RNA in ferroptosis of liver cancer and its impact on lipid peroxidation. Front Immunol 2025; 16:1555518. [PMID: 40207231 PMCID: PMC11979700 DOI: 10.3389/fimmu.2025.1555518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
Ferroptosis is an iron-dependent programmed death caused by the imbalance of lipid peroxides in cells. Unlike apoptosis, autophagy and necrosis, ferroptosis is mainly induced by the small molecule compound erastin. The main characteristics of ferroptosis were glutathione (GSH) depletion, inactivation of glutathione peroxidase 4 (GPX4) and reactive oxygen species (ROS) promoting lipid peroxidation. Eventually, the imbalance of lipid peroxidation regulation in cells leads to ferroptosis. The lipid metabolic pathway ultimately contributes to ferroptosis through the production of lipid peroxides. In addition, other cellular metabolic pathways can also regulate ferroptosis, such as the antioxidant metabolic pathway, which inhibits ferroptosis by clearing lipid peroxides and reducing cell membrane damage. Long non-coding RNAs (lncRNAs) are non-coding transcripts more than 200 nucleotides in length and are a less classified group of RNA transcripts that are associated with tumorigenesis and metastasis and are more tissue or cell type specific than protein-coding genes. Studies on the molecular profile of lncRNAs in plasma samples from liver cancer patients show that differentially expressed lncRNAs are mainly concentrated in biological functions related to tumorigenesis, such as cell metastasis, immune response and metabolic regulation. With different biological functions in physiological and pathological environments, the specific expression patterns of lncRNAs coordinate cell state, development, differentiation, and disease.
Collapse
Affiliation(s)
- Minglu Ding
- Graduate Student Department, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Keyuan Huo
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xiaojie Chen
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Wanyao Wang
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Zihan Xiang
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Yidan Song
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Peijian Chen
- College of Life Science, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Lantao Liu
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| |
Collapse
|