1
|
Mohamed HRH, Shaheen SEE, Ibrahim EH, Hussein NOE, Safwat G. Calcium titanate nanoparticles-induced cytotoxicity, genotoxicity and oxidative stress in human non-small lung cancer cells. Sci Rep 2025; 15:6373. [PMID: 39984527 PMCID: PMC11845463 DOI: 10.1038/s41598-025-89035-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/03/2025] [Indexed: 02/23/2025] Open
Abstract
Calcium titanate nanoparticles (CaTiO3NPs) have garnered significant attention due to their unique properties and excellent biocompatibility, which have led to their increased use in various fields and consumer products. This rise in application necessitates a better understanding of their biological and toxicological effects. However, there is limited data on the cytotoxicity and genotoxicity of CaTiO3NPs in human normal skin fibroblasts (HSF) and non-small lung cancer (A-549) cells. Consequently, this study aimed to explore the effect of 48-hour exposure to CaTiO3NPs on cell viability, genomic DNA integrity, and oxidative stress induction in human cancer A-549 cells, compared to normal HSF cells. The cytotoxicity and genotoxicity of CaTiO3NPs were assessed using the Sulforhodamine B (SRB) cytotoxicity and Alkaline Comet assays, respectively. To estimate possible oxidative stress induction and variation in apoptotic gene expression, reactive oxygen species (ROS) analysis and quantitative real-time polymerase chain reaction (qRT-PCR) were also performed. Our findings demonstrated that exposure to CaTiO3NPs for 48 h resulted in low toxicity toward both normal HSF and cancer A-549 cells, with cell death observed only at high concentrations (100 and 1000 µg/ml). The IC50 value of CaTiO3NPs in both HSF and A-549 cells was greater than 1000 µg/ml; specifically, the IC50 value in A-549 cells at 48 h was 1670.65 µg /ml. However, treatment with CaTiO3NPs for 48 h at the IC50 concentration of 1670.65 µg /ml resulted in significant genomic DNA damage and excessive ROS generation, along with a notable disturbance in the expression level of apoptotic (p53 and Bax) and anti-apoptotic Bcl2 genes in A-549 cells. In contrast, no significant changes were observed in HSF cells treated for 48 h with the same concentration (1670.65 µg /ml) of CaTiO3NPs. Collectively, these findings indicated that despite short-term exposure to CaTiO3NPs causing low cytotoxicity in both normal HSF and A-549 cells. CaTiO3NPs were selectively genotoxic toward A-549 cells. This genotoxicity was mediated through excessive ROS generation, which disrupted genomic DNA integrity and altered the expression of apoptotic genes, triggering apoptosis in A-549 cells. Further in vitro and in vivo studies are needed to fully understand the toxicological and biological properties of CaTiO3NPs.
Collapse
Affiliation(s)
- Hanan R H Mohamed
- Zoology Department Faculty of Science, Cairo University, Giza, Egypt.
| | - Shahd E E Shaheen
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
| | - Esraa H Ibrahim
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
| | - Nesma O E Hussein
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
| | - Gehan Safwat
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
| |
Collapse
|
2
|
Javar N, Sadat Shandiz SA, Abbasi M. Phyto-mediated fabrication of silver nanoparticles from Scrophularia striata extract (AgNPs-SSE): a potential inducer of apoptosis in breast cancer cells. Mol Biol Rep 2025; 52:172. [PMID: 39878805 DOI: 10.1007/s11033-025-10298-5] [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: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Breast carcinoma stands out as the most widespread invasive cancer and the top contributor to cancer-related mortality in women. Nanoparticles have emerged as promising tools in cancer detection, diagnosis, and prevention. In this study, the antitumor and apoptotic capability of silver nanoparticles synthesized through Scrophularia striata extract (AgNPs-SSE) was investigated toward breast cancer cells. METHODS The produced AgNPs-SSE were identified using scanning electron micrograph (SEM), energy-dispersive X-ray (EDAX) spectroscopy, Fourier-transform infrared (FTIR) spectroscopy, and X-ray diffraction (XRD). The cell-killing effects of AgNPs-SSE on MDA-MB231 mammary carcinoma cells were evaluated in vitro using the MTT assay over 24 h. Apoptosis induction was conducted by cell cycle analysis, annexin V-FITC/PI staining, reactive oxygen species (ROS) generation, and Hoechst staining. Additionally, the gene expression of βcatenin, GSK3β, and CyclinD1 was analyzed using quantitative real-time PCR (qRT-PCR). RESULTS Microscopic analysis confirmed the successful fabrication of globular AgNPs-SSE, with a mean particle dimension of 19 ± 10 nm. The MTT assay revealed that IC50 value of AgNPs-SSE was 48.5 µg/mL for mammary carcinoma cells and 114 µg/mL for normal cells. Annexin V-FITC/PI staining specified that 85.88% of cancer cells treated with AgNPs-SSE underwent either early or late apoptosis. Treatment with AgNPs-SSE also caused a considerable rise in the subG1 cell cycle population and ROS production. Furthermore, the upregulation of βcatenin and downregulation of CyclinD1 gene expression confirmed the apoptotic mechanism. CONCLUSIONS In conclusion, the findings suggest that phyto-synthesized AgNPs-SSE can restrain the expansion of breast carcinoma cells and provoke apoptosis through oxidative stress. These results highlight the potential of AgNPs-SSE as an antitumor agent against breast cancer.
Collapse
Affiliation(s)
- Niloufar Javar
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | - Maryam Abbasi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
3
|
Chaisupasakul P, Pekthong D, Wangteeraprasert A, Kaewkong W, Somran J, Kaewpaeng N, Parhira S, Srisawang P. Combination of ethyl acetate fraction from Calotropis gigantea stem bark and sorafenib induces apoptosis in HepG2 cells. PLoS One 2024; 19:e0300051. [PMID: 38527038 PMCID: PMC10962855 DOI: 10.1371/journal.pone.0300051] [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: 06/12/2023] [Accepted: 02/15/2024] [Indexed: 03/27/2024] Open
Abstract
The cytotoxicity of the ethyl acetate fraction of the Calotropis gigantea (L.) Dryand. (C. gigantea) stem bark extract (CGEtOAc) has been demonstrated in many types of cancers. This study examined the improved cancer therapeutic activity of sorafenib when combined with CGEtOAc in HepG2 cells. The cell viability and cell migration assays were applied in HepG2 cells treated with varying concentrations of CGEtOAc, sorafenib, and their combination. Flow cytometry was used to determine apoptosis, which corresponded with a decline in mitochondrial membrane potential and activation of DNA fragmentation. Reactive oxygen species (ROS) levels were assessed in combination with the expression of the phosphatidylinositol-3-kinase (PI3K)/ protein kinase B (Akt)/ mammalian target of rapamycin (mTOR) pathway, which was suggested for association with ROS-induced apoptosis. Combining CGEtOAc at 400 μg/mL with sorafenib at 4 μM, which were their respective half-IC50 concentrations, significantly inhibited HepG2 viability upon 24 h of exposure in comparison with the vehicle and each single treatment. Consequently, CGEtOAc when combined with sorafenib significantly diminished HepG2 migration and induced apoptosis through a mitochondrial-correlation mechanism. ROS production was speculated to be the primary mechanism of stimulating apoptosis in HepG2 cells after exposure to a combination of CGEtOAc and sorafenib, in association with PI3K/Akt/mTOR pathway suppression. Our results present valuable knowledge to support the development of anticancer regimens derived from the CGEtOAc with the chemotherapeutic agent sorafenib, both of which were administered at half-IC50, which may minimize the toxic implications of cancer treatments while improving the therapeutic effectiveness toward future medical applications.
Collapse
Affiliation(s)
- Pattaraporn Chaisupasakul
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
| | - Dumrongsak Pekthong
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand
- Center of Excellence for Environmental Health and Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand
| | | | - Worasak Kaewkong
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Julintorn Somran
- Department of Pathology, Faculty of Medicine, Naresuan University, Phitsanulok, Thailand
| | - Naphat Kaewpaeng
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand
| | - Supawadee Parhira
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
- Center of Excellence for Environmental Health and Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand
| | - Piyarat Srisawang
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, Thailand
- Center of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
4
|
Jayaraman S, Veeraraghavan VP, Natarajan SR, Jasmine S. Exploring the therapeutic potential of curcumin in oral squamous cell carcinoma (HSC-3 cells): Molecular insights into hypoxia-mediated angiogenesis. Pathol Res Pract 2024; 254:155130. [PMID: 38277750 DOI: 10.1016/j.prp.2024.155130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Oral cancer represents a substantial global health burden, often associate with hypoxia-induced angiogenesis as a critical factor in its progression. Curcumin, a naturally occurring bioactive compounds, has gained increasing attention for its potential anticancer properties. OBJECTIVE To assess the impact of curcumin on oral cancer, particularly its role in modulating HIF-1α-mediated angiogenesis in HSC-3 cells. METHODS Our investigation involved multiple experimental approaches, including MTT assay, aerobic glycolysis by metabolic kit, cell cycle, and apoptosis assessment via flow cytometry. Furthermore, we employed molecular docking techniques to examine the interactions between curcumin and key angiogenesis related proteins, including HIF-1α, VEGF-B, MMP-3, and STAT3. RESULTS Our results demonstrate that curcumin exerts significant effects on the cell survivability, cell cycle regulation, and apoptosis induction in oral cancer cells. These effects were particularly pronounced under the conditions of HIF-1α mediated angiogenesis. Computational binding analysis revealed strong binding interactions with curcumin and the selected proteins, implying a plausible mechanism through which curcumin may modulate the angiogenic pathways in oral cancer. CONCLUSION Our research sheds light on the diverse effects of curcumin on oral cancer cells, emphasizing its potential as a promising therapeutic tool for addressing hypoxia-induced angiogenesis. However, further investigation is essential to comprehensively understand the molecular mechanisms underlying these effects in in vitro models. This deeper comprehension is crucial for translating these findings into clinical applications aimed at improving oral cancer treatment.
Collapse
Affiliation(s)
- Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, SaveethaUniversity, Chennai 600 077, India
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, SaveethaUniversity, Chennai 600 077, India.
| | - Sathan Raj Natarajan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, SaveethaUniversity, Chennai 600 077, India
| | - Sharmila Jasmine
- Department of Oral Maxillofacial Surgery, Rajas Dental College and Hospital, Kavalkinaru, Tirunelveli 627105, Tamil Nadu, India
| |
Collapse
|
5
|
Jayaraman S, Natararaj S, Veeraraghavan VP. Hesperidin Inhibits Oral Cancer Cell Growth via Apoptosis and Inflammatory Signaling-Mediated Mechanisms: Evidence From In Vitro and In Silico Analyses. Cureus 2024; 16:e53458. [PMID: 38435153 PMCID: PMC10909395 DOI: 10.7759/cureus.53458] [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: 11/28/2023] [Accepted: 02/02/2024] [Indexed: 03/05/2024] Open
Abstract
Background Oral carcinoma presents a significant health challenge, prompting the need for innovative therapeutic approaches. Elevation of inflammatory mediators, including tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), has promoted cellular proliferation, inhibited apoptosis, and fostered oral cancer progression through complex signaling pathways. Hesperidin, a flavanone glycoside found in citrus fruits, is of keen interest in this study as it has been proven to have multiple health benefits through in vivo and in vitro studies. However, the mechanism behind the anticancer activity of hesperidin in oral carcinoma remains obscure. Aim The study aimed to explore the anticancer potential of hesperidin on human oral cancer cells (KB cells) by modulating pro-inflammatory and apoptotic signaling mechanisms. Methods Cancer cell growth inhibitory activity was assessed using the MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) assay. Gene expression analysis was performed using real-time RT-PCR analysis. In addition, in silico docking analysis was conducted to confirm the binding affinity of hesperidin with pro-inflammatory and apoptosis signaling molecules. The data were analyzed using one-way ANOVA and the "t" test. Results Utilizing the MTT assay, a dose-dependent cytotoxic effect of hesperidin was unveiled, with a remarkable IC50 value indicative of its potent inhibition of cell proliferation. Complementing these findings (p<0.05), qRT-PCR analysis demonstrated hesperidin's regulatory influence on key molecular targets within the KB cell line. Hesperidin treatment resulted in a noteworthy reduction in TNF-α, interleukin-1 beta (IL-1-β), IL-6, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and B-cell lymphoma 2 (Bcl-2) mRNA expression levels (p<0.05), highlighting its inhibitory role in cell proliferation, migration, and inflammation processes. Simultaneously, hesperidin promoted the expression of BAX mRNA (p<0.05), indicating an enhancement in cell death. Molecular docking simulations further revealed robust binding affinities between hesperidin and target proteins, suggesting its potential to disrupt cellular functions and inflammatory signaling pathways in oral cancer cells. Conclusion The cytotoxic effects on the KB cell line and its anti-inflammatory properties position hesperidin as a compelling candidate for further exploration in the quest for effective oral carcinoma treatments. These findings shed light on the intricate molecular mechanisms underlying hesperidin's promise as a therapeutic agent against oral carcinoma.
Collapse
Affiliation(s)
- Selvaraj Jayaraman
- Department of Biochemistry, Centre of Molecular Medicine and Diagnostics (COMManD) Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Sathanraj Natararaj
- Department of Biochemistry, Centre of Molecular Medicine and Diagnostics (COMManD) Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Centre of Molecular Medicine and Diagnostics (COMManD) Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|