1
|
Eskandari A, Safavi SN, Sahrayi H, Alizadegan D, Eskandarisani M, Javanmard A, Tajik M, Sadeghi Z, Toutounch A, Yeganeh FE, Noorbazargan H. Antimicrobial and antibiofilm activity of prepared thymol@UIO-66 and thymol/ZnONPs@UIO-66 nanoparticles against Methicillin-resistant Staphylococcus aureus: A synergistic approach. Colloids Surf B Biointerfaces 2025; 249:114529. [PMID: 39879671 DOI: 10.1016/j.colsurfb.2025.114529] [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: 10/10/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/31/2025]
Abstract
This study introduces a novel approach to enhance the antibacterial properties of UIO-66 by incorporating both Thymol and ZnO nanoparticles within its framework which represents a significant advancement like exhibiting a synergistic antibacterial effect, providing a prolonged and controlled release, and mitigating cytotoxicity associated with the release of free ZnO nanoparticles by combining these two antimicrobial agents within a single, well-defined metal-organic framework. UIO-66 frameworks are investigated as carriers for the natural antimicrobial agent, Thymol, and ZnONPs offering a novel drug delivery system for antibacterial applications. Results demonstrated 132, 90, 184, and 223 nm sizes for UIO-66, ZnONPs, UIO-66 encapsulated Thymol, and UIO-66 encapsulated both Thymol and ZnONPs, respectively. Successful encapsulation of the antibacterial drug with a high entrapment efficiency of 64 % for Thymol was approved, and 49 % in-vitro release of Thymol was achieved for 72 hours. In-vitro antibacterial assays revealed promising results, with the drug-loaded nanoparticles exhibiting significantly lower MIC values and enhanced bactericidal activity against S. Aureus bacterial strains compared to the free drug, as demonstrated by agar disk diffusion and time-kill assays. MIC values reduced from a range of 31.25-250 µg/ml for free Thymol and 12.5-100 µg/ml for free ZnONPs to 3.9-62.5 µg/ml for Thymol@UIO-66 and 1.95-15.63 µg/ml for Thymol/ZnONPs@UIO-66. According to the results, the mixture of both Thymol and ZnONPs had 41 % and 16 % more antibiofilm activities in comparison with free Thymol and free ZnONPs, respectively. Furthermore, Thymol@UIO-66 had 25 % higher antibiofilm activities relative to not-encapsulated Thymol and ZnONPs, and this improvement was even 46 % more in Thymol/ZnONPs@UIO-66 in comparison with Thymol@UIO-66. Overall, this study demonstrates the potential of Thymol/ZnONPs@UIO-66 frameworks as a promising drug delivery platform for effective antibacterial therapy. This approach to overcome antibiotic resistance and improve treatment efficacy potentially.
Collapse
Affiliation(s)
- Alireza Eskandari
- CTERC, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Nooshin Safavi
- Department of Polymer Engineering, Faculty of Engineering, Qom University of Technology, Qom, Iran
| | - Hamidreza Sahrayi
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Dorsa Alizadegan
- Faculty of Pharmacy, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran
| | | | - Alireza Javanmard
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA 16802-1503, United States
| | - Mohammadreza Tajik
- Biomedical Engineering Department, Carnegie Mellon University, Pittsburgh, PA 15219, United States
| | - Zohre Sadeghi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Disease, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Arvin Toutounch
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Hassan Noorbazargan
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Jiang W, Zhang Y, Wang Q. Exploring the molecular mechanisms network of breast cancer by multi-omics analysis. Asia Pac J Clin Oncol 2025; 21:129-137. [PMID: 38477438 PMCID: PMC11733836 DOI: 10.1111/ajco.14052] [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: 07/06/2023] [Revised: 01/07/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND Breast cancer (BC), the most prevalent malignancy in women globally, still lacks comprehensive research on its molecular targets and necessitates further investigation into the underlying molecular mechanisms driving its initiation and progression. METHODS The GSE20685 Series Matrix File downloaded from the Gene Expression Omnibus database was divided into a high-risk group (n = 49) and a low-risk group (n = 278) to construct the co-expression network. RESULTS Four hub genes were identified based on the Weighted Gene Co-expression Network Analysis. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analyses were performed. Hub gene immune infiltration was investigated using the Tumor Immune Estimation Resource database, and CD4+ T cell expression levels were substantially correlated with hub gene expression. Based on the CancerRxGene database (Genomics of Drug Sensitivity in Cancer database), it was found that the hub genes were highly sensitive to common chemotherapy drugs such as AKT inhibitor VIII and Erlotinib. The expression of Secreted Frizzled-Related Protein 1, melanoma-inhibiting activity (MIA), and Keratin 14 was related to tumor mutation burden, and the expression of MIA also affected the microsatellite instability of the tumor. This study employs multi-omics analysis to investigate the molecular network associated with the prognosis of BC, highlighting its intricate connection with the immune microenvironment. CONCLUSION These findings pinpoint four crucial genes in BC progression, offering targets for further research and therapy. Their connections to immune infiltration and chemotherapy sensitivity underscore complex interactions in the tumor microenvironment.
Collapse
Affiliation(s)
- Wei Jiang
- Department of AnesthesiologyYongchuan Hospital of Chongqing Medical UniversityChongqingChina
| | - Yanjun Zhang
- Department of Breast SurgeryYongchuan Hospital of Chongqing Medical UniversityChongqingChina
| | - Qiuqiong Wang
- Department of Respiratory and Critical Care MedicineYongchuan Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
3
|
Chen X, Wu W, Jeong JH, Rokavec M, Wei R, Feng S, Schroth W, Brauch H, Zhong S, Luo JL. Cytokines-activated nuclear IKKα-FAT10 pathway induces breast cancer tamoxifen-resistance. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1413-1426. [PMID: 38565741 DOI: 10.1007/s11427-023-2460-0] [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: 06/05/2023] [Accepted: 09/26/2023] [Indexed: 04/04/2024]
Abstract
Endocrine therapy that blocks estrogen signaling is the most effective treatment for patients with estrogen receptor positive (ER+) breast cancer. However, the efficacy of agents such as tamoxifen (Tam) is often compromised by the development of resistance. Here we report that cytokines-activated nuclear IKKα confers Tam resistance to ER+ breast cancer by inducing the expression of FAT10, and that the expression of FAT10 and nuclear IKKα in primary ER+ human breast cancer was correlated with lymphotoxin β (LTB) expression and significantly associated with relapse and metastasis in patients treated with adjuvant mono-Tam. IKKα activation or enforced FAT10 expression promotes Tam-resistance while loss of IKKα or FAT10 augments Tam sensitivity. The induction of FAT10 by IKKα is mediated by the transcription factor Pax5, and coordinated via an IKKα-p53-miR-23a circuit in which activation of IKKα attenuates p53-directed repression of FAT10. Thus, our findings establish IKKα-to-FAT10 pathway as a new therapeutic target for the treatment of Tam-resistant ER+ breast cancer.
Collapse
Affiliation(s)
- Xueyan Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Weilin Wu
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Ji-Hak Jeong
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Matjaz Rokavec
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Rui Wei
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shaolong Feng
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Werner Schroth
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, 70376, Germany
- iFIT Cluster of Excellence, University of Tübingen, Tübingen, 72074, Germany
| | - Hiltrud Brauch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, 70376, Germany
- iFIT Cluster of Excellence, University of Tübingen, Tübingen, 72074, Germany
| | - Shangwei Zhong
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA.
- The Cancer Research Institute and the Second Affiliated Hospital, Henyang Medical School, University of South China, Hengyang, 421001, China.
| | - Jun-Li Luo
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA.
- The Cancer Research Institute and the Second Affiliated Hospital, Henyang Medical School, University of South China, Hengyang, 421001, China.
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China.
| |
Collapse
|
4
|
Motafeghi F, Mortazavi P, Shokrzadeh M. Anticancer activity of zinc oxide nanoparticles on prostate and colon cancer cell line. Toxicol Res (Camb) 2024; 13:tfad127. [PMID: 38239270 PMCID: PMC10793725 DOI: 10.1093/toxres/tfad127] [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: 05/18/2023] [Revised: 11/27/2023] [Accepted: 12/16/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Considering the numerous drug resistance in cancer and the advancement of science in nanomedicines, it was decided to compare the effectiveness of zinc oxide nanoparticles in colon and prostate cell lines. Considering the importance of factors and Oxidative stress pathways in cancer prevention, the aim of the study is based on oxidative stress mechanisms. Methodes In order to evaluate the effects of zinc oxide nanoparticles on colon and prostate cell lines, oxidative stress factors ROS, MDA, and GSH and mitochondrial function were evaluated. The data was analyzed with Prism v8 software, and the significance level was considered to be P < 0.05. Results The results showed that nanoparticles induce ROS and reduce intracellular glutathione by destroying and disrupting mitochondrial function, and by increasing ROS production, damage to the lipid membrane and an increase in MDA were also evident. This effect was dose-dependent and the greatest at a concentration of 25 μg/mL. Also, ZnO nanoparticles performed better in the HT29 cell line than in the PC3 cell line. Conclusion This study showed that exposure of HT29 and PC3 cancer cells to zinc oxide nanoparticles at different concentrations inhibited growth by cytotoxic effects.
Collapse
Affiliation(s)
- Farzaneh Motafeghi
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences and Metabolism, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Parham Mortazavi
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 1583-88994, Iran
| | - Mohammad Shokrzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
5
|
Lin Y, Deng H, Deng F, Yao S, Deng X, Cheng Y, Chen Y, He B, Dai W, Zhang H, Zhang Q, Wang X. Remodeling of intestinal epithelium derived extracellular vesicles by nanoparticles and its bioeffect on tumor cell migration. J Control Release 2024; 365:60-73. [PMID: 37972765 DOI: 10.1016/j.jconrel.2023.11.011] [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: 07/15/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Extracellular vesicles (EVs) are an effective tool to elucidate the bioeffect of nanomedicines. To clarify the interaction between oral nanomedicines and intestinal epithelial cells, and their bioeffects on downstream cells, polystyrene nanoparticles (PS-NPs) with different sizes were used as the model nanomedicines for EVs induction. Caco-2 monolayers were selected as the model of the intestinal epithelium and DLD-1 cells as the colorectal cancer model proximal to the gastrointestinal tract. It is found that compared with small-sized (25, 50, 100 nm) PS-NPs, the large-sized (200 and 500 nm) exhibited higher co-localization with multivesicular bodies and lysosomes, and more significant reduction of lysosomal acidification in Caco-2 cells. Proteomic and western-blotting analysis showed that the EVs remodeled by large-sized PS-NPs exhibited a higher extent of protein expression changes. The in vitro and in vivo signaling pathway detection in DLD-1 cells and DLD-1 cell xenograft nude mice showed that the remodeled EVs by large-sized PS-NPs inhibited the activation of multiple signaling pathways including Notch3, EGF/EGFR, and PI3K/Akt pathways, which resulted in the inhibition of tumor cell migration. These results primarily clarify the regulation mechanisms of nanomedicines-EVs-receptor cells chain. It provides a new perspective for the rational design and bioeffect evaluation of oral drug nanomaterials and sets up the fundamental knowledge for novel tumor therapeutics in the future.
Collapse
Affiliation(s)
- Yuxing Lin
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hailiang Deng
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Feiyang Deng
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA 02215, USA
| | - Siyu Yao
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xinxin Deng
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuxi Cheng
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ying Chen
- Guangdong Institute for Drug Control, Guangzhou 510700, China; NMPA Key Laboratory for Quality Control and Evaluation of Pharmaceutical Excipients, Guangzhou 510700, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; NMPA Key Laboratory for Quality Control and Evaluation of Pharmaceutical Excipients, Guangzhou 510700, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; NMPA Key Laboratory for Quality Control and Evaluation of Pharmaceutical Excipients, Guangzhou 510700, China.
| |
Collapse
|
6
|
Yeh YS, Tsai HL, Chen PJ, Chen YC, Su WC, Chang TK, Huang CW, Wang JY. Identifying and clinically validating biomarkers for immunotherapy in colorectal cancer. Expert Rev Mol Diagn 2023; 23:231-241. [PMID: 36908268 DOI: 10.1080/14737159.2023.2188195] [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: 12/06/2022] [Accepted: 03/03/2023] [Indexed: 03/14/2023]
Abstract
INTRODUCTION Colorectal cancer (CRC) is a leading cause of death. For three decades, chemotherapy with or without targeted therapy (provided before or after tumor resection surgery) has been the standard treatment for patients with CRC. Biomarkers are key tools for performing early detection, prognostication, and survival and treatment response predictions. Notably, immune checkpoint inhibitors (ICIs) have transformed prognoses for solid tumors (including CRC). AREA COVERED Although immunotherapy has developed considerably, it is only effective for a small number of microsatellite instability-high (MSIH) cancer cases; such cases represent only 5% of metastatic CRC (mCRC) cases, which are characterized by an immune-inflamed microenvironment that can be rewired against cancer cells through ICI administration. Immunotherapy research is gradually uncovering the mechanism underlying immune resistance in patients with CRC and discovering new biomarkers. For example, studies have clinically validated the associations of deficient mismatch repair system/microsatellite instability, tumor mutation burden, programmed death ligand 1 expression, and polymerase epsilon with CRC in patients undergoing immunotherapy. EXPERT OPINIONS Clinical trials documenting the effect of immune checkpoints were performed to produce long-lasting effects for patients with mCRC. Consequently, therapeutic decision-making models are further refined by the inclusion of powerful molecular biomarkers in patients with CRC.
Collapse
Affiliation(s)
- Yung-Sung Yeh
- Division of Trauma and Surgical Critical Care, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Emergency Medicine, Faculty of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Hsiang-Lin Tsai
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Jung Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Chih Su
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung-Kun Chang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, Faculty of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Wen Huang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jaw-Yuan Wang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Pingtung Hospital, Ministry of Health and Welfare, Pingtung, Taiwan
| |
Collapse
|
7
|
Hannachi E, Khan FA, Slimani Y, Rehman S, Trabelsi Z, Akhtar S, Al-Suhaimi EA. In Vitro Antimicrobial and Anticancer Peculiarities of Ytterbium and Cerium Co-Doped Zinc Oxide Nanoparticles. BIOLOGY 2022; 11:1836. [PMID: 36552345 PMCID: PMC9775757 DOI: 10.3390/biology11121836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Zinc oxide nanoparticles (ZnO NPs) are a promising platform for their use in biomedical research, especially given their anticancer and antimicrobial activities. This work presents the synthesis of ZnO NPs doped with different amounts of rare-earth ions of ytterbium (Yb) and cerium (Ce) and the assessment of their anticancer and antimicrobial activities. The structural investigations indicated a hexagonal wurtzite structure for all prepared NPs. The particle size was reduced by raising the amount of Ce and Yb in ZnO. The anticancer capabilities of the samples were examined by the cell viability MTT assay. Post 48-h treatment showed a reduction in the cancer cell viability, which was x = 0.00 (68%), x = 0.01 (58.70%), x = 0.03 (80.94%) and x = 0.05 (64.91%), respectively. We found that samples doped with x = 0.01 and x = 0.05 of Yb and Ce showed a better inhibitory effect on HCT-116 cancer cells than unadded ZnO (x = 0.00). The IC50 for HCT-116 cells of Ce and Yb co-doped ZnO nanoparticles was calculated and the IC50 values were x = 0.01 (3.50 µg/mL), x = 0.05 (8.25 µg/mL), x = 0.00 (11.75 µg/mL), and x = 0.03 (21.50 µg/mL). The treatment-doped ZnO NPs caused apoptotic cell death in the HCT-116 cells. The nanoparticles showed inhibitory action on both C. albicans and E. coli. It can be concluded that doping ZnO NPs with Yb and Ce improves their apoptotic effects on cancer and microbial cells.
Collapse
Affiliation(s)
- Essia Hannachi
- Department of Nuclear Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Firdos Alam Khan
- Department of Stem Cell Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Yassine Slimani
- Department of Biophysics, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Suriya Rehman
- Department of Epidemic Diseases Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Zayneb Trabelsi
- Department of Physics, Faculty of Science of Bizerte, University of Carthage, Bizerte 7021, Tunisia
| | - Sultan Akhtar
- Department of Biophysics, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Ebtesam A. Al-Suhaimi
- Biology Department & Institute for Research and Medical Consultations (IRMC), College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
8
|
Tang S, Ling Z, Jiang J, Gu X, Leng Y, Wei C, Cheng H, Li X. Integrating the tumor-suppressive activity of Maspin with p53 in retuning the epithelial homeostasis: A working hypothesis and applicable prospects. Front Oncol 2022; 12:1037794. [PMID: 36523976 PMCID: PMC9745138 DOI: 10.3389/fonc.2022.1037794] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/10/2022] [Indexed: 12/20/2024] Open
Abstract
Epithelial malignant transformation and tumorous development were believed to be closely associated with the loss of its microenvironment integrity and homeostasis. The tumor-suppressive molecules Maspin and p53 were demonstrated to play a crucial role in body epithelial and immune homeostasis. Downregulation of Maspin and mutation of p53 were frequently associated with malignant transformation and poor prognosis in various human cancers. In this review, we focused on summarizing the progress of the molecular network of Maspin in studying epithelial tumorous development and its response to clinic treatment and try to clarify the underlying antitumor mechanism. Notably, Maspin expression was reported to be transcriptionally activated by p53, and the transcriptional activity of p53 was demonstrated to be enhanced by its acetylation through inhibition of HDAC1. As an endogenous inhibitor of HDAC1, Maspin possibly potentiates the transcriptional activity of p53 by acetylating the p53 protein. Hereby, it could form a "self-propelling" antitumor mechanism. Thus, we summarized that, upon stimulation of cellular stress and by integrating with p53, the aroused Maspin played the epigenetic surveillant role to prevent the epithelial digressional process and retune the epithelial homeostasis, which is involved in activating host immune surveillance, regulating the inflammatory factors, and fine-tuning its associated cell signaling pathways. Consequentially, in a normal physiological condition, activation of the above "self-propelling" antitumor mechanism of Maspin and p53 could reduce cellular stress (e.g., chronic infection/inflammation, oxidative stress, transformation) effectively and achieve cancer prevention. Meanwhile, designing a strategy of mimicking Maspin's epigenetic regulation activity with integrating p53 tumor-suppressive activity could enhance the chemotherapy efficacy theoretically in a pathological condition of cancer.
Collapse
Affiliation(s)
- Sijie Tang
- Department of Urology, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, Suzhou, China
- Aoyang Cancer Institute, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, Suzhou, China
| | - Zhongli Ling
- Department of Urology, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, Suzhou, China
| | - Jiajia Jiang
- Aoyang Cancer Institute, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, Suzhou, China
| | - Xiang Gu
- Department of Urology, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, Suzhou, China
| | - Yuzhong Leng
- Department of Urology, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, Suzhou, China
| | - Chaohui Wei
- Department of Urology, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, Suzhou, China
| | - Huiying Cheng
- Aoyang Cancer Institute, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, Suzhou, China
| | - Xiaohua Li
- Aoyang Cancer Institute, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, Suzhou, China
| |
Collapse
|
9
|
Pehlivanoglu S, Acar CA, Donmez S. Characterization of green synthesized flaxseed zinc oxide nanoparticles and their cytotoxic, apoptotic and antimigratory activities on aggressive human cancer cells. INORG NANO-MET CHEM 2021. [DOI: 10.1080/24701556.2021.1980034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Suray Pehlivanoglu
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, Konya, Turkey
| | - Cigdem Aydin Acar
- Bucak School of Health, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Soner Donmez
- Bucak School of Health, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
10
|
Gurzu S, Jung I. Subcellular Expression of Maspin in Colorectal Cancer: Friend or Foe. Cancers (Basel) 2021; 13:366. [PMID: 33498377 PMCID: PMC7864036 DOI: 10.3390/cancers13030366] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 02/05/2023] Open
Abstract
In this review the authors aimed to emphasize the practical value of nuclear expression of the mammary serine protease inhibitor (maspin), also known as serpin B5 protein, in colorectal carcinoma (CRC), from pre-malignant disorders to carcinogenesis and metastasis. As the role of maspin is controversial and not yet understood, the present update highlights the latest data revealed by literature which were filtrated through the daily experience of the authors, which was gained at microscopic examination of maspin expression in CRCs and other tumors for daily diagnosis. Data regarding the subcellular localization of maspin, in correlation with the microsatellite status, grade of tumor dedifferentiation, and epithelial-mesenchymal transition (EMT) phenomenon of the tumor buds were presented with details. An original observation refers to the maspin capacity to mark the tumor cells which are "at the point of budding" that were previously considered as having "hybrid EMT phenotype". It refers to the transitional status of tumor cell that is between "epithelial status" and "mesenchymal status". The second original hypothesis highlights the possible role of maspin in dysregulating the intestinal microbiota, in patients with idiopathic inflammatory bowel diseases (IBD) and inducing IBD-related CRC. The dynamic process of budding and EMT of tumor buds, possible mediated by maspin, needs further investigation and validation in many human CRC samples. The histological and molecular data reveal that synthesis of maspin-based therapeutics might represent a novel individualized therapeutic strategy for patients with CRC.
Collapse
Affiliation(s)
- Simona Gurzu
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, 540139 Targu-Mures, Romania;
| | | |
Collapse
|
11
|
Ahamed M, Akhtar MJ, Khan MAM, Alhadlaq HA. SnO 2-Doped ZnO/Reduced Graphene Oxide Nanocomposites: Synthesis, Characterization, and Improved Anticancer Activity via Oxidative Stress Pathway. Int J Nanomedicine 2021; 16:89-104. [PMID: 33447029 PMCID: PMC7802795 DOI: 10.2147/ijn.s285392] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/30/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Therapeutic selectivity and drug resistance are critical issues in cancer therapy. Currently, zinc oxide nanoparticles (ZnO NPs) hold considerable promise to tackle this problem due to their tunable physicochemical properties. This work was designed to prepare SnO2-doped ZnO NPs/reduced graphene oxide nanocomposites (SnO2-ZnO/rGO NCs) with enhanced anticancer activity and better biocompatibility than those of pure ZnO NPs. MATERIALS AND METHODS Pure ZnO NPs, SnO2-doped ZnO (SnO2-ZnO) NPs, and SnO2-ZnO/rGO NCs were prepared via a facile hydrothermal method. Prepared samples were characterized by field emission transmission electron microscopy (FETEM), energy dispersive spectroscopy (EDS), field emission scanning electron microscopy (FESEM), X-ray diffraction (XRD), ultraviolet-visible (UV-VIS) spectrometer, and dynamic light scattering (DLS) techniques. Selectivity and anticancer activity of prepared samples were assessed in human breast cancer (MCF-7) and human normal breast epithelial (MCF10A) cells. Possible mechanisms of anticancer activity of prepared samples were explored through oxidative stress pathway. RESULTS XRD spectra of SnO2-ZnO/rGO NCs confirmed the formation of single-phase of hexagonal wurtzite ZnO. High resolution TEM and SEM mapping showed homogenous distribution of SnO2 and rGO in ZnO NPs with high quality lattice fringes without any distortion. Band gap energy of SnO2-ZnO/rGO NCs was lower compared to SnO2-ZnO NPs and pure ZnO NPs. The SnO2-ZnO/rGO NCs exhibited significantly higher anticancer activity against MCF-7 cancer cells than those of SnO2-ZnO NPs and ZnO NPs. The SnO2-ZnO/rGO NCs induced apoptotic response through the upregulation of caspase-3 gene and depletion of mitochondrial membrane potential. Mechanistic study indicated that SnO2-ZnO/rGO NCs kill cancer cells through oxidative stress pathway. Moreover, biocompatibility of SnO2-ZnO/rGO NCs was also higher against normal breast epithelial (MCF10A cells) in comparison to SnO2-ZnO NPs and ZnO NPs. CONCLUSION SnO2-ZnO/rGO NCs showed enhanced anticancer activity and better biocompatibility than SnO2-ZnO NPs and pure ZnO NPs. This work suggested a new approach to improve the selectivity and anticancer activity of ZnO NPs. Studies on antitumor activity of SnO2-ZnO/rGO NCs in animal models are further warranted.
Collapse
Affiliation(s)
- Maqusood Ahamed
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh11451, Saudi Arabia
| | - Mohd Javed Akhtar
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh11451, Saudi Arabia
| | - M A Majeed Khan
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh11451, Saudi Arabia
| | - Hisham A Alhadlaq
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh11451, Saudi Arabia
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh11451, Saudi Arabia
| |
Collapse
|