1
|
Miao H, Zhang B, Li Y, Ma X, Yang Y, Lin Z, Liu Y. Rosuvastatin inhibits carcinogenesis through Ca 2+ triggered endoplasmic reticulum stress pathway in pancreatic cancer. Cell Signal 2025; 131:111753. [PMID: 40107481 DOI: 10.1016/j.cellsig.2025.111753] [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: 01/01/2025] [Revised: 02/17/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Pancreatic cancer remains one of the most challenging malignancies to treat due to its late-stage diagnosis, aggressive progression, and high resistance to existing therapies. Rosuvastatin (ROV), known for its hypolipidemic effects, which significantly inhibited clonogenic capacity and epithelial-mesenchymal transition (EMT) in prostate cancer cells. However, the anti-cancer mechanisms of ROV in PC have not yet been fully explored. PURPOSE This study aimed to investigate the potential anti-cancer effects of ROV on PC cells and to elucidate the underlying mechanisms. METHODS Cytotoxicity was detected via MTT assay, while epithelial-mesenchymal transition (EMT) markers, Ca2+ levels, and endoplasmic reticulum (ER) stress were observed with fluorescence microscopy. RNA-seq analysis was used to identify significantly changed mRNA expression following ROV treatment. Additionally, western blotting and immunohistochemistry (IHC) were conducted to examine proteins involving in the cell cycle, EMT, Ca2+ signaling, and endoplasmic reticulum stress (ERS) in vitro and in vivo. RESULTS ROV inhibited PC cell proliferation by arresting the cell cycle at the G1/S phase and partially reducing cell mobility during the EMT process. A total of 1336 significantly different RNAs (P < 0.05 and |logFC|>1) were identified and analyzed through RNA-seq, revealing the Ca2+ and ER pathways in PC cells treated with ROV. ROV treatment significantly altered the level of intracellular Ca2+, triggering the ERS pathway and modulating the Ca2+/CaM/CaMKII/ERK pathway. Furthermore, ROV inhibited key proteins within the Ca2+ and ERS pathways, leading to reduced cell proliferation, mobility and G1/S phase arrest. In tumor tissues, the expression of Ki67, EMT markers, Calmodulin, and ATF6 corroborated the in vitro findings. CONCLUSION ROV inhibited proliferation and metastasis in PC cells by inhibiting the EMT process through the Ca2+/CaM/CaMKII/ERK and Ca2+-mediated ERS pathways, highlighting its potential as a prophylactic and therapeutic agent for PC.
Collapse
Affiliation(s)
- Hui Miao
- Central Laboratory, Yanbian University Hospital, Yanji 133000, China; Dunhua City Hospital, Dunhua 133700, China
| | - Baojian Zhang
- Central Laboratory, Yanbian University Hospital, Yanji 133000, China; Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji 133002, China
| | - Yue Li
- Central Laboratory, Yanbian University Hospital, Yanji 133000, China; Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji 133002, China
| | - Xiao Ma
- Central Laboratory, Yanbian University Hospital, Yanji 133000, China; Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji 133002, China
| | - Yang Yang
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji 133002, China
| | - Zhenhua Lin
- Central Laboratory, Yanbian University Hospital, Yanji 133000, China; Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji 133002, China
| | - Yanqun Liu
- Central Laboratory, Yanbian University Hospital, Yanji 133000, China; Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji 133002, China.
| |
Collapse
|
2
|
Dang Y, Zhang Y, Wang Z. The role of statins in the regulation of breast and colorectal cancer and future directions. Front Pharmacol 2025; 16:1578345. [PMID: 40438592 PMCID: PMC12116307 DOI: 10.3389/fphar.2025.1578345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/08/2025] [Indexed: 06/01/2025] Open
Abstract
Statins, widely recognized as a cornerstone in the prevention of cardiovascular diseases, have garnered increasing attention in oncology due to their pleiotropic effects, particularly their potential roles in regulating breast and colorectal cancer. Emerging evidence suggests that statins may exert anticancer effects through multiple mechanisms, including the mitochondrial apoptosis pathway, the LKB1-AMPK-p38MAPK-p53-survivin signaling cascade, inhibition of the mevalonate pathway, modulation of the EGFR/RhoA and IGF-1 signaling pathways, and regulation of the BMP/SMAD4 signaling pathway. However, significant heterogeneity exists in the reported anticancer effects of statins, likely due to variations in statin type (lipophilic vs hydrophilic), dosage, treatment duration, and population-specific characteristics. These factors contribute to inconsistencies in study outcomes. Additionally, while combination therapies incorporating statins with chemotherapy and immunotherapy have demonstrated synergistic effects in certain studies, their clinical utility remains to be fully established. Nevertheless, current evidence suggests that statins may have a potential role in reducing breast and colorectal cancer-related mortality. Future research should prioritize elucidating their precise molecular mechanisms, defining dose-response relationships, developing personalized treatment strategies within the framework of precision medicine, and validating their efficacy through large-scale, long-term prospective studies. These efforts will provide a more robust scientific foundation for the clinical application of statins in oncology. This review systematically explores the role of statins in breast and colorectal cancer regulation, covering clinical evidence, underlying biological mechanisms, pharmacological distinctions, synergistic therapeutic potential, and translational medicine prospects.
Collapse
Affiliation(s)
| | | | - Zhihao Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Kumari R, Banerjee S. Regulation of Different Types of Cell Death by Noncoding RNAs: Molecular Insights and Therapeutic Implications. ACS Pharmacol Transl Sci 2025; 8:1205-1226. [PMID: 40370994 PMCID: PMC12070317 DOI: 10.1021/acsptsci.4c00681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
Noncoding RNAs (ncRNAs) are crucial regulatory molecules in various biological processes, despite not coding for proteins. ncRNAs are further divided into long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) based on the size of their nucleotides. These ncRNAs play crucial roles in transcriptional, post-transcriptional, and epigenetic regulation. The regulatory roles of noncoding RNAs, including lncRNAs, miRNAs, and circRNAs, are essential in various modalities of cellular death, such as apoptosis, ferroptosis, cuproptosis, pyroptosis, disulfidptosis, and necroptosis. These noncoding RNAs are integral to modulating gene expression and protein functionality during cellular death mechanisms. In apoptosis, lncRNAs, miRNAs, and circRNAs influence the transcription of apoptotic genes. In ferroptosis, these noncoding RNAs target genes and proteins involved in iron homeostasis and oxidative stress responses. For cuproptosis, noncoding RNAs regulate pathways associated with the accumulation of copper ions, leading to cellular death. During pyroptosis, noncoding RNAs modulate inflammatory mediators and caspases, affecting the proinflammatory cell death pathway. In necroptosis, noncoding RNAs oversee the formation and functionality of necrosomes, thereby influencing the balance between cellular survival and death. Disulfidptosis is a unique type of regulated cell death caused by the excessive formation of disulfide bonds within cells, leading to cytoskeletal collapse and oxidative stress, especially under glucose-limited conditions. This investigation highlights the complex mechanisms through which noncoding RNAs coordinate cellular death, emphasizing their therapeutic promise as potential targets, particularly in the domain of cancer treatment.
Collapse
Affiliation(s)
- Reshmi Kumari
- Department of Biotechnology, School
of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Satarupa Banerjee
- Department of Biotechnology, School
of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
4
|
Espejo EF, Guerra MDM, Castellano S. Association between serum myeloperoxidase enzyme activity and Parkinson's disease status. NPJ Parkinsons Dis 2025; 11:94. [PMID: 40287421 PMCID: PMC12033337 DOI: 10.1038/s41531-025-00941-0] [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: 10/25/2024] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Elevated levels of the inflammatory enzyme myeloperoxidase (MPO) in the blood are associated with the development of age-related inflammatory diseases. Given that age, inflammation, and blood MPO play a role in the pathogenesis of Parkinson´s disease (PD), we hypothesized that serum MPO could be associated with PD status. This case-control study (199 participants) was conducted using an extensive protocol, and the concentration and activity of MPO in blood serum were measured. The findings reveal that serum MPO concentration and activity are significantly increased in the patients, and that rates of PD in all individuals are associated with increasing tertiles of MPO concentration and activity. In multivariate logistic regression model adjusting for confounding factors, MPO activity (not concentration) is the factor that is most associated with PD status (OR, 6.921, P = 0.001). Mental depression is directly associated with MPO activity and with PD status (OR, 0.121, B = -2.108, P = 0.002). The use of statins or nonsteroidal anti-inflammatory drugs significantly reduces serum MPO activity, but the possible association with the odds of having PD does not survive correction for multiple testing. In summary, both serum MPO concentration and activity are increased in patients with PD, but only MPO enzyme activity is associated with PD status. These findings may have implications for the evaluation of PD.
Collapse
Affiliation(s)
| | - María-Del-Mar Guerra
- Department of Biochemistry, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Silvia Castellano
- Department of Metabolopathy, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
5
|
Tan LK, Liu J, Ma CZ, Huang S, He FH, Long Y, Zheng ZS, Liang JL, Xu N, Wang G, Liu YF. Iron-Dependent Cell Death: Exploring Ferroptosis as a Unique Target in Triple-Negative Breast Cancer Management. Cancer Manag Res 2025; 17:625-637. [PMID: 40124838 PMCID: PMC11930262 DOI: 10.2147/cmar.s503932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/25/2025] [Indexed: 03/25/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by aggressive behavior, high metastatic potential, and frequent relapses, presenting significant treatment challenges. Ferroptosis, a unique form of programmed cell death marked by iron-dependent lipid peroxidation, has emerged as a crucial factor in cancer biology. Recent studies indicate that TNBC cells possess a distinct metabolic profile linked to iron and glutathione, which may render them more susceptible to ferroptosis than other breast cancer subtypes. Moreover, ferroptosis plays a role in the interactions between immune cells and tumor cells, suggesting its potential to modulate the tumor microenvironment and influence the immune response against TNBC.Evidence reveals that ferroptosis not only affects TNBC cell viability but also alters the tumor microenvironment by promoting the release of damage-associated molecular patterns (DAMPs), which can recruit immune cells to the tumor site. Specific ferroptosis-related genes and biomarkers, such as ACSL4 and GPX4, demonstrate altered expression patterns in TNBC tissues, offering promising avenues for diagnostic and prognostic applications. Furthermore, in preclinical models, the induction of ferroptosis has been shown to enhance the efficacy of existing therapies, indicating a synergistic effect that could be harnessed for therapeutic benefit. The compelling link between ferroptosis and TNBC underscores its potential as a novel therapeutic target. Future research should focus on developing strategies that exploit ferroptosis in conjunction with traditional therapies, including the identification of natural compounds and efficacious ferroptosis inducers for personalized treatment regimens. This review elucidates the multifaceted implications of ferroptosis in TNBC, providing valuable insights for improving both diagnosis and treatment of this formidable breast cancer subtype.
Collapse
Affiliation(s)
- Li-kuan Tan
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| | - Jiaxing Liu
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| | - Cheng-zhi Ma
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| | - Shaolong Huang
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| | - Feng-hui He
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| | - Yang Long
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| | - Zhi-sheng Zheng
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| | - Jia-liang Liang
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| | - Nan Xu
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| | - Guanghui Wang
- Breast Surgery, Guizhou Provincial People’s Hospital, Guiyang, People’s Republic of China
| | - Yu-fei Liu
- Breast Surgery, Tongren People’s Hospital, Tongren, People’s Republic of China
| |
Collapse
|
6
|
Du J, Meng X, Yang M, Chen G, Li J, Zhu Z, Wu X, Hu W, Tian M, Li T, Ren S, Zhao P. NGR-Modified CAF-Derived exos Targeting Tumor Vasculature to Induce Ferroptosis and Overcome Chemoresistance in Osteosarcoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410918. [PMID: 39889249 PMCID: PMC11948032 DOI: 10.1002/advs.202410918] [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] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/13/2024] [Indexed: 02/02/2025]
Abstract
Osteosarcoma (OS) chemoresistance presents a significant clinical challenge. This study aims to investigate the potential of using tumor vascular-targeting peptide NGR-modified cancer-associated fibroblasts (CAFs)-derived exosomes (exos) to deliver circ_0004872-encoded small peptides promoting autophagy-dependent ferroptosis to reverse chemoresistance in OS. Through combined single-cell transcriptome analysis and high-throughput sequencing, it identified circ_0004872 associated with chemoresistance. Subsequent experiments demonstrated that the small peptide encoded by this Circular RNA (circRNA) can effectively reverse chemoresistance by enhancing OS cell sensitivity to chemotherapy via the mechanism of promoting autophagy-dependent ferroptosis. Moreover, in vitro and in vivo results confirmed the efficient delivery of NGR-modified CAFs-derived exo-packaged circ_0004872-109aa to tumor cells, thereby improving targeted therapy efficacy. This study not only offers a novel strategy to overcome chemoresistance in OS but also highlights the potential application value of utilizing exos for drug delivery.
Collapse
Affiliation(s)
- Jianxin Du
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Xiangwei Meng
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Minghao Yang
- Department of RadiologyYantai Affiliated Hospital of Binzhou Medical UniversityYantai264100China
| | - Guancheng Chen
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
| | - Jigang Li
- Department of OrthopedicsZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Zengjun Zhu
- School of Medical LaboratoryShandong Second Medical UniversityWeifang261042China
| | - Xuanxuan Wu
- School of Medical LaboratoryShandong Second Medical UniversityWeifang261042China
| | - Wei Hu
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Maojin Tian
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Tao Li
- Department of OrthopedicsNanjing Jiangbei HospitalNanjing210044China
| | - Shuai Ren
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Peiqing Zhao
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| |
Collapse
|
7
|
Atale N, Wells A. Statins as Secondary Preventive Agent to Limit Breast Cancer Metastatic Outgrowth. Int J Mol Sci 2025; 26:1300. [PMID: 39941069 PMCID: PMC11818786 DOI: 10.3390/ijms26031300] [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: 12/22/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Metastasis is a leading cause of mortality in breast cancer, as metastatic disease is often aggressive and resistant to conventional treatments. Cancer cells that spread to distant organs can enter a dormant phase for extended periods, sometimes years or decades. During this dormant phase, cancer cells avoid immune and pharmacological response. Thus, new approaches are needed to prevent these disseminated cells from becoming lethal cancers. Statins are known inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase that have been extensively used in patients with cardiovascular diseases to lower cholesterol. However, recent research has demonstrated their potential in anticancer therapies. Epidemiological evidence suggests that statins are associated with a reduction in breast cancer-specific mortality, although they do not appear to affect the incidence of primary tumors. In this review, we discuss the role of statins in metastasis and dormancy, their cytocidal and cytostatic effects and their interactions with different cell types in the tumor microenvironment. The exact mechanisms by which statins reduce mortality without influencing primary tumor growth remain unclear, also warranting further investigation into their potential role in metastasis and tumor dormancy, which could ultimately help patients to improve survival and quality of life.
Collapse
Affiliation(s)
- Neha Atale
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Alan Wells
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Research and Development Service, Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
- Cell Biology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
8
|
Liu J, Tang R, Zheng J, Luo K. Targeting ferroptosis reveals a new strategy for breast cancer treatment: a bibliometric study. Discov Oncol 2024; 15:679. [PMID: 39560863 DOI: 10.1007/s12672-024-01569-x] [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: 09/19/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Studies exploring the role of ferroptosis in the pathogenesis of breast cancer have proliferated over the past decade, especially in 2023, with a staggering 217 publications in related studies. However, there are still significant gaps in comprehensive scientometric analysis and mapping of scientific studies, especially in terms of temporal and study area tracking, principal investigators, and the emergence of new hotspots. OBJECTIVE This study aims to summarize the role of ferroptosis in the development of breast cancer and the latest research results on the ferroptosis-targeted treatment of breast cancer and to use bibliometric methods to draw a visual map to explore future research trends. METHODS On May 11, 2024, this study updated the research progress related to ferroptosis and breast cancer over the past 11 years by retrieving data from January 1, 2014, to May 1, 2024, from the Web of Science database. In this research, many scientific analysis software including VOSviewer, chorddiag R Language Pack, Scimago Graphica, Citespace 6.3.R1, Cluster Profiler, enrichplot, ggplot2 R Language Pack, Cytoscape, and STRING online platform are used to make in-depth scientific analysis and visualization of the measurement results. RESULTS Statistical analysis of these data showed that China accounted for 74.43% of the total publications, highlighting China's dominant role in research on the relationship between ferroptosis and breast cancer. Several research institutions, including Sun Yat-sen University, Zhejiang University, and Shanghai Jiao Tong University, have achieved impressive results. Efferth, Thomas is the most prominent author in this field and has the highest number of publications in the subfield of oncology. This study clearly shows that ferroptosis plays a crucial role in the development of triple-negative breast cancer, hepatocellular carcinoma, glioma, leukemia, mitochondrial disease, lymphoma, bladder tumors, lung adenocarcinoma, and esophageal tumors. CONCLUSION This study provides a comprehensive bibliometric evaluation that deepens our understanding of the role of ferroptosis in the pathogenesis of breast cancer and the current status of targeting ferroptosis for treating breast cancer. Thus, it helps researchers in related fields explore new research directions by comprehensively extracting important information and research hotspots.
Collapse
Affiliation(s)
- Junlin Liu
- Pathology Department of Jingmen Central Hospital, Jingmen, Hubei, China
- Pathology Department of Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| | - Rong Tang
- Pathology Department of Jingmen Central Hospital, Jingmen, Hubei, China
- Pathology Department of Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| | - Jie Zheng
- Pathology Department of Jingmen Central Hospital, Jingmen, Hubei, China
- Pathology Department of Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| | - Ke Luo
- Pathology Department of Jingmen Central Hospital, Jingmen, Hubei, China.
- Pathology Department of Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, Hubei, China.
| |
Collapse
|
9
|
Pańczyszyn E, Lallukka M, Gagliardi M, Saverio V, Monzani R, Miola M, Verné E, Corazzari M. Tellurium-Doped Bioactive Glass Induces Ferroptosis in Osteosarcoma Cells Regardless of FSP1. Antioxidants (Basel) 2024; 13:1327. [PMID: 39594469 PMCID: PMC11591201 DOI: 10.3390/antiox13111327] [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: 09/17/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Human osteosarcoma (OS) is a rare tumor predominantly affecting long bones and characterized by a poor prognosis. Currently, the first line of intervention consists of the surgical resection of primary tumors combined with radiotherapy and chemotherapy, with a profound impact on the patient's life. Since the surgical removal of OS frequently results in a large resection of bones, the use of biomaterials to sustain the stability of the remaining tissue and to stimulate bone regeneration is challenging. Moreover, residual neoplastic cells might be responsible for tumor recurrence. Here, we explored the potential of tellurium-ion-doped bioactive glass as a novel therapeutic intervention to both eradicate residual malignant cells and promote bone regeneration. Bioactive glass (BAG) has been extensively studied and employed in the field of regenerative medicine due to its osseointegration properties and ability to improve bone tissue regeneration. We found that the incorporation of tellurium (Te) in BAG selectively kills OS cells through ferroptosis while preserving the viability of hBMSCs and stimulating their osteodifferentiation. However, the mechanism of Te toxicity is still unclear: (i) Te-BAG generates lipid-ROS through LOXs activity but not iron overload; (ii) Te-dependent ferroptosis is mediated by GPX4 down-regulation; and (iii) the anti-ferroptotic activity of FSP1 is abrogated, whose expression confers the resistance of OS to the canonical induction of ferroptosis. Overall, our data show that Te-doped bioglass could represent an interesting biomaterial with both pro-ferroptotic activity towards residual cancer cells and pro-osteoregenerative activity.
Collapse
Affiliation(s)
- Elżbieta Pańczyszyn
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.P.); (M.G.); (V.S.); (R.M.)
| | - Mari Lallukka
- Applied Science and Technology Department, Politecnico di Torino, 10129 Turin, Italy; (M.L.); (M.M.); (E.V.)
| | - Mara Gagliardi
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.P.); (M.G.); (V.S.); (R.M.)
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Valentina Saverio
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.P.); (M.G.); (V.S.); (R.M.)
| | - Romina Monzani
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.P.); (M.G.); (V.S.); (R.M.)
| | - Marta Miola
- Applied Science and Technology Department, Politecnico di Torino, 10129 Turin, Italy; (M.L.); (M.M.); (E.V.)
| | - Enrica Verné
- Applied Science and Technology Department, Politecnico di Torino, 10129 Turin, Italy; (M.L.); (M.M.); (E.V.)
| | - Marco Corazzari
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.P.); (M.G.); (V.S.); (R.M.)
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
10
|
Chen YH, Wu JX, Yang SF, Wu YC, Hsiao YH. Molecular Mechanisms Underlying the Anticancer Properties of Pitavastatin against Cervical Cancer Cells. Int J Mol Sci 2024; 25:7915. [PMID: 39063157 PMCID: PMC11277542 DOI: 10.3390/ijms25147915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/27/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Cervical cancer ranks as the fourth most prevalent form of cancer and is a significant contributor to female mortality on a global scale. Pitavastatin is an anti-hyperlipidemic medication and has been demonstrated to exert anticancer and anti-inflammatory effects. Thus, the purpose of this study was to evaluate the anticancer effect of pitavastatin on cervical cancer and the underlying molecular mechanisms involved. The results showed that pitavastatin significantly inhibited cell viability by targeting cell-cycle arrest and apoptosis in Ca Ski, HeLa and C-33 A cells. Pitavastatin caused sub-G1- and G0/G1-phase arrest in Ca Ski and HeLa cells and sub-G1- and G2/M-phase arrest in C-33 A cells. Moreover, pitavastatin induced apoptosis via the activation of poly-ADP-ribose polymerase (PARP), Bax and cleaved caspase 3; inactivated the expression of Bcl-2; and increased mitochondrial membrane depolarization. Furthermore, pitavastatin induced apoptosis and slowed the migration of all three cervical cell lines, mediated by the PI3K/AKT and MAPK (JNK, p38 and ERK1/2) pathways. Pitavastatin markedly inhibited tumor growth in vivo in a cancer cell-originated xenograft mouse model. Overall, our results identified pitavastatin as an anticancer agent for cervical cancer, which might be expanded to clinical use in the future.
Collapse
Affiliation(s)
- Ya-Hui Chen
- Women’s Health Research Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan; (Y.-H.C.); (J.-X.W.)
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
| | - Jyun-Xue Wu
- Women’s Health Research Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan; (Y.-H.C.); (J.-X.W.)
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Yun-Chia Wu
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua 50006, Taiwan;
| | - Yi-Hsuan Hsiao
- Women’s Health Research Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan; (Y.-H.C.); (J.-X.W.)
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua 50006, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|