1
|
Xiang Z, Ma B, Pei X, Wang W, Gong W. Mechanism of action of genistein on breast cancer and differential effects of different age stages. PHARMACEUTICAL BIOLOGY 2025; 63:141-155. [PMID: 39996512 PMCID: PMC11864014 DOI: 10.1080/13880209.2025.2469607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
CONTEXT Genistein, a soy-derived isoflavone, exhibits structural similarities with 17β-estradiol and demonstrates antioxidant, anti-inflammatory, and estrogenic properties. Despite its low bioavailability limiting its clinical application, it shows potential for breast cancer prevention and treatment. OBJECTIVE This review aims to summarize the pharmacological effects and molecular mechanisms of genistein in breast cancer, focusing on its therapeutic potential, strategies to overcome bioavailability limitations, and its role in personalized medicine. Differential impacts among population subgroups are also discussed. METHODS A systematic review was conducted using PubMed, ScienceDirect, and Google Scholar databases. Studies were selected based on their focus on genistein's mechanisms of action, strategies to enhance its bioavailability, and interactions with other therapies. RESULTS Genistein exerted anticancer effects by modulating estrogen receptor β (ERβ), inhibiting angiogenesis, arresting the cell cycle, and inducing apoptosis. Its antioxidant properties help mitigate tumor-associated oxidative stress. Bioavailability enhancement strategies, such as nanoparticle and lipid-based formulations, show promise. Age-dependent effects were evident, with distinct responses observed in prepubertal, menopausal, and postmenopausal populations, underscoring its potential for personalized therapies. Furthermore, genistein influences epigenetic modifications, including DNA methylation and miRNA expression, bolstering its anticancer efficacy. CONCLUSION Genistein is a promising candidate for breast cancer therapy, particularly for personalized treatment. Strategies to enhance bioavailability and further clinical research are essential to optimize its therapeutic potential and evaluate its efficacy in combination therapies.
Collapse
Affiliation(s)
- Zhebin Xiang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Ma
- Zhejiang Hospital, Hangzhou, China
| | - Xiujun Pei
- Shandong Provincial Hospital, Shandong, China
| | - Wenjie Wang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Weilun Gong
- Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Ur Rahman M, Hussain HR, Akram H, Sarfraz M, Nouman M, Khan JA, Ishtiaq M. Niosomes as a targeted drug delivery system in the treatment of breast cancer: preparation, classification and mechanisms of cellular uptake. J Drug Target 2025; 33:916-932. [PMID: 39964023 DOI: 10.1080/1061186x.2025.2468750] [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: 09/27/2024] [Revised: 01/24/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
Breast cancer (BC) remains one of the significant health issues across the globe, being diagnosed in millions of women worldwide annually. Conventional therapeutic options have substantial adverse effects due to their non-specificity and limited drug bioavailability. Niosomes, being novel drug delivery systems formed from non-ionic surfactants, with or without cholesterol and charge-inducing agents, are used as therapeutic options in treating BC. Their formulation by various methods enhances the therapeutic efficacy and bioavailability and minimises side effects. Niosomal formulation of tamoxifen exhibits target drug delivery with enhanced stability, whereas docetaxel and methotrexate show sustained and controlled drug release, respectively. 5-Fluorouracil, doxorubicin, paclitaxel, cyclophosphamide and epirubicin show improved cytotoxic effects against BC when combined with other agents. Furthermore, repurposed niosomal formulations of anti-cancer drugs show improved penetration, reduced tumour volume and significantly enhanced anti-tumour effect. This review article focuses on the composition of niosomes and their application in BC treatment and then examines how niosomes could contribute to BC research.
Collapse
Affiliation(s)
| | | | - Habiba Akram
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Sarfraz
- College of Pharmacy, Al-Ain University, Al-Ain, United Arab Emirates
| | - Muhammad Nouman
- College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Jawad Akbar Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore, Pakistan
| | - Memona Ishtiaq
- Department of Pharmacy, Lahore Institute of Professional Studies, Lahore, Pakistan
| |
Collapse
|
3
|
Mansouri N, Daneshgar M, Khojasteh F, Modaresi Z, Taheri R, Mokaberi P, Saberi MR, Chamani J. RAS/RAF/MEK/MAPK signaling pathway as a therapeutic target in breast cancer: Emphasis on a novel carrier for tamoxifen and digestion behaviors. AVICENNA JOURNAL OF PHYTOMEDICINE 2025; 15:1204-1227. [PMID: 40365182 PMCID: PMC12068497 DOI: 10.22038/ajp.2024.25253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/08/2024] [Indexed: 05/15/2025]
Abstract
Objective This research attempted to increase the bioactivity and solubility and reduce the side effects of Tamoxifen (TMX) by using the cellulose nanocrystals (CNCs) extracted from walnut shells as a carrier and studied the interaction behavior of CNCs-TMX with hemoglobin. Materials and Methods The synthesized CNCs and CNCs-TMX were analyzed through the usage of XRD, FTIR, TEM, SEM, and multi-spectroscopic techniques. A real-time PCR assay was also conducted to further unravel the underlying mechanism of CNCs- TMX. Results Our synthesized products including CNCs and CNCs- TMX had spherical morphologies in small sizes of 17.42 nm and 56.38 nm, respectively. The changes in FTIR spectrum signified the induced alterations in the samples functional group during the steps of preparation, while the crystallinity index of CNCs was 71.35%. Fluorescence spectroscopy confirmed the quencher functionality of CNCs-TMX along with the dominance of static quenching mechanism. Also, synchronous fluorescence displayed its binding to Hb in the vicinity of Tryptophanresidue. FRET was applied to calculate the interaction energy transfer of 0.18 nm. Next to achieving satisfactory results from oxygen-hemoglobin dissociation studies, the presence of CNCs-TMX caused a reduction in hemoglobin affinity for oxygen. Conclusion Our findings pointed out the remarkable potential of TMX-loaded CNCs, derived from walnut shell, in suppressing the proliferation, migration, and invasion of breast cancer cells by quelling the RAS/RAF/MEK/MAPK signaling pathways. The gathered data approved the promising applicability of the obtained CNCs from walnut shell in the delivery system of anti-cancer drugs throughout pharmaceutical applications.
Collapse
Affiliation(s)
- Niloofar Mansouri
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Melika Daneshgar
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Farzaneh Khojasteh
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Zahra Modaresi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Reza Taheri
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Parisa Mokaberi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Mohammad Reza Saberi
- DepartmentofMedicinalChemistry,SchoolofPharmacy,MashhadUniversityofMedicalSciences,Mashhad,Iran
| | - Jamshidkhan Chamani
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
4
|
Cheng Z, Huang H, Yin M, Liu H. Applications of liposomes and lipid nanoparticles in cancer therapy: current advances and prospects. Exp Hematol Oncol 2025; 14:11. [PMID: 39891180 PMCID: PMC11786384 DOI: 10.1186/s40164-025-00602-1] [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] [Received: 11/20/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025] Open
Abstract
Liposomes and lipid nanoparticles are common lipid-based drug delivery systems and play important roles in cancer treatment and vaccine manufacture. Although significant progress has been made with these lipid-based nanocarriers in recent years, efficient clinical translation of active targeted liposomal nanocarriers remains extremely challenging. In this review, we focus on targeted liposomes, stimuli-responsive strategy and combined therapy in cancer treatment. We also summarize advances of liposome and lipid nanoparticle applications in nucleic acid delivery and tumor vaccination. In addition, we discuss limitations and challenges in the clinical translation of these lipid nanomaterials and make recommendations for the future research in cancer therapy.
Collapse
Affiliation(s)
- Zhe Cheng
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Huichao Huang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Infectious Disease, XiangYa Hospital, Central South University, Changsha, 410008, China
| | - Meilong Yin
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Huaizheng Liu
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
5
|
Beram FM, Ali SN, Mesbahian G, Pashizeh F, Keshvadi M, Mashayekhi F, Khodadadi B, Bashiri Z, Moeinzadeh A, Rezaei N, Namazifard S, Hossein-Khannazer N, Tavakkoli Yaraki M. 3D Printing of Alginate/Chitosan-Based Scaffold Empowered by Tyrosol-Loaded Niosome for Wound Healing Applications: In Vitro and In Vivo Performances. ACS APPLIED BIO MATERIALS 2024; 7:1449-1468. [PMID: 38442406 DOI: 10.1021/acsabm.3c00814] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
This study introduces a tyrosol-loaded niosome integrated into a chitosan-alginate scaffold (Nio-Tyro@CS-AL), employing advanced electrospinning and 3D printing techniques for wound healing applications. The niosomes, measuring 185.40 ± 6.40 nm with a polydispersity index of 0.168 ± 0.012, encapsulated tyrosol with an efficiency of 77.54 ± 1.25%. The scaffold's microsized porous structure (600-900 μm) enhances water absorption, promoting cell adhesion, migration, and proliferation. Mechanical property assessments revealed the scaffold's enhanced resilience, with niosomes increasing the compressive strength, modulus, and strain to failure, indicative of its suitability for wound healing. Controlled tyrosol release was demonstrated in vitro, essential for therapeutic efficacy. The scaffold exhibited significant antibacterial activity against Pseudomonas aeruginosa and Staphylococcus aureus, with substantial biofilm inhibition and downregulation of bacterial genes (ndvb and icab). A wound healing assay highlighted a notable increase in MMP-2 and MMP-9 mRNA expression and the wound closure area (69.35 ± 2.21%) in HFF cells treated with Nio-Tyro@CS-AL. In vivo studies in mice confirmed the scaffold's biocompatibility, showing no significant inflammatory response, hypertrophic scarring, or foreign body reaction. Histological evaluations revealed increased fibroblast and macrophage activity, enhanced re-epithelialization, and angiogenesis in wounds treated with Nio-Tyro@CS-AL, indicating effective tissue integration and repair. Overall, the Nio-Tyro@CS-AL scaffold presents a significant advancement in wound-healing materials, combining antibacterial properties with enhanced tissue regeneration, and holds promising potential for clinical applications in wound management.
Collapse
Affiliation(s)
| | - Saba Naeimaei Ali
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin 3419759811, Iran
| | - Ghazal Mesbahian
- School of Pharmacy, International Campus, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Fatemeh Pashizeh
- Department of Immunology, School of Medicine, Shahid Sadoughi University of Medical Science, Yazd 8916188635, Iran
| | | | - Farzaneh Mashayekhi
- Rasoul Akram Hospital, Iran University of Medical Sciences (IUMS), Tehran 14535, Iran
| | - Behnoosh Khodadadi
- Department of Chemistry, Faculty of Physics and Chemistry, Alzahra University, Tehran 1993891176, Iran
| | - Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Alaa Moeinzadeh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Niloufar Rezaei
- Gastroenterology and Liver Diseases Research Center, Research, Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| | - Saina Namazifard
- Department of Mechanical and Aerospace Engineering, The University of Texas at Arlington, 500 West First Street, Arlington, Texas 76019, United States
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research, Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| | - Mohammad Tavakkoli Yaraki
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
6
|
Liga S, Paul C, Moacă EA, Péter F. Niosomes: Composition, Formulation Techniques, and Recent Progress as Delivery Systems in Cancer Therapy. Pharmaceutics 2024; 16:223. [PMID: 38399277 PMCID: PMC10892933 DOI: 10.3390/pharmaceutics16020223] [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: 12/04/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Niosomes are vesicular nanocarriers, biodegradable, relatively non-toxic, stable, and inexpensive, that provide an alternative for lipid-solid carriers (e.g., liposomes). Niosomes may resolve issues related to the instability, fast degradation, bioavailability, and insolubility of different drugs or natural compounds. Niosomes can be very efficient potential systems for the specific delivery of anticancer, antioxidant, anti-inflammatory, antimicrobial, and antibacterial molecules. This review aims to present an overview of their composition, the most common formulation techniques, as well as of recent utilizations as delivery systems in cancer therapy.
Collapse
Affiliation(s)
- Sergio Liga
- Biocatalysis Group, Department of Applied Chemistry and Engineering of Organic and Natural Compounds, Faculty of Industrial Chemistry and Environmental Engineering, Politehnica University Timișoara, Carol Telbisz 6, 300001 Timișoara, Romania; (S.L.); (F.P.)
| | - Cristina Paul
- Biocatalysis Group, Department of Applied Chemistry and Engineering of Organic and Natural Compounds, Faculty of Industrial Chemistry and Environmental Engineering, Politehnica University Timișoara, Carol Telbisz 6, 300001 Timișoara, Romania; (S.L.); (F.P.)
| | - Elena-Alina Moacă
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2nd Eftimie Murgu Square, 300041 Timișoara, Romania;
| | - Francisc Péter
- Biocatalysis Group, Department of Applied Chemistry and Engineering of Organic and Natural Compounds, Faculty of Industrial Chemistry and Environmental Engineering, Politehnica University Timișoara, Carol Telbisz 6, 300001 Timișoara, Romania; (S.L.); (F.P.)
- Research Institute for Renewable Energies, Politehnica University Timișoara, Gavril Muzicescu 138, 300501 Timișoara, Romania
| |
Collapse
|
7
|
Pashizeh F, Mansouri A, Bazzazan S, Abdihaji M, Khaleghian M, Bazzazan S, Rezei N, Eskandari A, Mashayekhi F, Heydari M, Tavakkoli Yaraki M. Bioresponsive gingerol-loaded alginate-coated niosomal nanoparticles for targeting intracellular bacteria and cancer cells. Int J Biol Macromol 2024; 258:128957. [PMID: 38154726 DOI: 10.1016/j.ijbiomac.2023.128957] [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: 09/15/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
Targeting and treating intracellular pathogen infections has been long-standing challenge, particularly in light of the escalating prevalence of antimicrobial resistance. Herein, an optimum formulation of alginate (AL)-coated niosome-based carriers for delivery of herbal extract Gingerol (Gin) was developed to treat intracellular pathogen infections and cancer cells. We used Gin-Nio@AL as a model drug to assess its efficacy against Gram-negative/positive bacteria and breast cancer cell lines. Our investigation affirmed its heightened antibacterial and anticancer properties. The antibacterial activity of Gin-Nio@AL against intracellular Staphylococcus aureus (S. aureus) and pseudomonas aeruginosa (P. aeruginosa) was also tested. In the current study, the niosome nanoparticles containing herbal extract Gingerol were optimized regarding lipid content and Surfactant per Cholesterol molar ratio. The developed formulation provided potential advantages, such as smooth globular surface morphology, small diameter (240.68 nm), pH-sensitive sustained release, and high entrapment efficiency (94.85 %). The release rate of Gin from AL-coated niosomes (Gin-Nio@AL) in physiological and acidic pH is lower than uncoated nanoparticles (Gin-Nio). Besides, the release rate of Gin from niosomal formulations increased in acidic pH. The Gin-Nio@AL demonstrated good antimicrobial activity against S. aureus and P. aeruginosa, and compared to Gin-Nio, the MIC values decreased to 7.82 ± 0.00 and 1.95 ± 0.00 μg/mL, respectively. In addition, the time-kill assay results showed that the developed formulation significantly reduced the number of bacteria in both strains compared to other tested groups. The microtiter data and scanning electron microscope micrography showed that Gin-Nio@AL has a more significant inhibitory effect on biofilm formation than Gin-Nio and Gin. The cell cytotoxicity evaluation showed that Gin-Nio@AL reduced the survival rate of MDA-MB-231 cancer cells to 52.4 % and 45.2 % after 48 h and 72 h, respectively. The elimination of intracellular pathogens was investigated through a breast cancer cell infection in an in vitro model. Gin-Nio@AL exhibited an enhanced and sustained intracellular antibacterial activity against pathogens-infected breast cancer cells compared to other tested formulations. Overall, Gin-Nio@AL enables the triggered release and targeting of intra-extra cellular bacteria and cancer cells and provides a novel and promising candidate for treating intracellular pathogen infections and cancer cells.
Collapse
Affiliation(s)
- Fatemeh Pashizeh
- Department of Immunology, School of Medicine, Shahid Sadoughi University of Medical Science Yazd, Iran
| | - Afsoun Mansouri
- School of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saina Bazzazan
- Department of Community Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Mohammadreza Abdihaji
- Department of Biology, The Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN, USA
| | | | - Saba Bazzazan
- Department of Community Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Niloufar Rezei
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Alireza Eskandari
- CTERC, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzaneh Mashayekhi
- Rasoul Akram Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Heydari
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Tavakkoli Yaraki
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
8
|
Zaer M, Moeinzadeh A, Abolhassani H, Rostami N, Tavakkoli Yaraki M, Seyedi SA, Nabipoorashrafi SA, Bashiri Z, Moeinabadi-Bidgoli K, Moradbeygi F, Farmani AR, Hossein-Khannazer N. Doxorubicin-loaded Niosomes functionalized with gelatine and alginate as pH-responsive drug delivery system: A 3D printing approach. Int J Biol Macromol 2023; 253:126808. [PMID: 37689301 DOI: 10.1016/j.ijbiomac.2023.126808] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/27/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Despite many efforts, breast cancer remains one of the deadliest cancers and its treatment faces challenges related to cancer drug side effects and metastasis. Combining 3D printing and nanocarriers has created new opportunities in cancer treatment. In this work, 3D-printed gelatin-alginate nanocomposites containing doxorubicin-loaded niosomes (Nio-DOX@GT-AL) were recruited as an advanced potential pH-sensitive drug delivery system. Morphology, degradation, drug release, flow cytometry, cell cytotoxicity, cell migration, caspase activity, and gene expression of nanocomposites and controls (Nio-DOX and Free-DOX) were evaluated. Results show that the obtained niosome has a spherical shape and size of 60-80 nm. Sustained drug release and biodegradability were presented by Nio-DOX@GT-AL and Nio-DOX. Cytotoxicity analysis revealed that the engineered Nio-DOX@GT-AL scaffold had 90 % cytotoxicity against breast cancer cells (MCF-7), whereas exhibited <5 % cytotoxicity against the non-tumor breast cell line (MCF-10A), which was significantly more than the antitumor effect of the control samples. Scratch-assay as an indicator cell migration demonstrated a reduction of almost 60 % of the covered surface. Gene expression could provide an explanation for the antitumor effect of engineered nanocarriers, which significantly reduced metastasis-promoting genes (Bcl2, MMP-2, and MMP-9), and significantly enhanced the expression and activity of genes that promote apoptosis (CASP-3, CASP-8, and CASP-9). Also, considerable inhibition of metastasis-associated genes (Bax and p53) was observed. Moreover, flow-cytometry data demonstrated that Nio-DOX@GT-AL decreased necrosis and enhanced apoptosis drastically. The findings of this research can confirm that employing 3D-printing and niosomal formulation can be an effective strategy in designing novel nanocarriers for efficient drug delivery applications.
Collapse
Affiliation(s)
- Mohammad Zaer
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Alaa Moeinzadeh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Abolhassani
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Neda Rostami
- Department of Chemistry, Amirkabir University of Technology, Tehran, Iran
| | - Mohammad Tavakkoli Yaraki
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| | - Seyed Arsalan Seyedi
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran, Iran
| | - Seyed Ali Nabipoorashrafi
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran, Iran
| | - Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kasra Moeinabadi-Bidgoli
- Basic and Molecular Epidemiology of Gastroenterology Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moradbeygi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Reza Farmani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research, Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Taghavizadeh Yazdi ME, Qayoomian M, Beigoli S, Boskabady MH. Recent advances in nanoparticle applications in respiratory disorders: a review. Front Pharmacol 2023; 14:1059343. [PMID: 37538179 PMCID: PMC10395100 DOI: 10.3389/fphar.2023.1059343] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 05/30/2023] [Indexed: 08/05/2023] Open
Abstract
Various nanoparticles are used in the discovery of new nanomedicine to overcome the shortages of conventional drugs. Therefore, this article presents a comprehensive and up-to-date review of the effects of nanoparticle-based drugs in the treatment of respiratory disorders, including both basic and clinical studies. Databases, including PubMed, Web of Knowledge, and Scopus, were searched until the end of August 2022 regarding the effect of nanoparticles on respiratory diseases. As a new tool, nanomedicine offered promising applications for the treatment of pulmonary diseases. The basic composition and intrinsic characteristics of nanomaterials showed their effectiveness in treating pulmonary diseases. The efficiency of different nanomedicines has been demonstrated in experimental animal models of asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer, lung infection, and other lung disorders, confirming their function in the improvement of respiratory disorders. Various types of nanomaterials, such as carbon nanotubes, dendrimers, polymeric nanomaterials, liposomes, quantum dots, and metal and metal oxide nanoparticles, have demonstrated therapeutic effects on respiratory disorders, which may lead to new possible remedies for various respiratory illnesses that could increase drug efficacy and decrease side effects.
Collapse
Affiliation(s)
| | - Mohsen Qayoomian
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sima Beigoli
- Mashhad University of Medical Sciences, Mashhad, Razavi Khorasan, Iran
| | - Mohammad Hossein Boskabady
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Zhao W, Chang Y, Wu Z, Jiang X, Li Y, Xie R, Fu D, Sun C, Gao J. Identification of PIMREG as a novel prognostic signature in breast cancer via integrated bioinformatics analysis and experimental validation. PeerJ 2023; 11:e15703. [PMID: 37483962 PMCID: PMC10358341 DOI: 10.7717/peerj.15703] [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: 03/19/2023] [Accepted: 06/14/2023] [Indexed: 07/25/2023] Open
Abstract
Background Phosphatidylinositol binding clathrin assembly protein interacting mitotic regulator (PIMREG) expression is upregulated in a variety of cancers. However, its potential role in breast cancer (BC) remains uncertain. Methods The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were used to gather relevant information. The expression of PIMREG and its clinical implication in BC were assessed by using Wilcoxon rank-sum test. The prognostic value of PIMREG in BC was evaluated through the Cox regression model and nomogram, and visualized by Kaplan-Meier survival curves. Genes/proteins that interact with PIMREG in BC were also identified through GeneMANIA and MaxLink. Gene set enrichment analysis (GSEA) was then performed. The correlations of the immune cell infiltration and immune checkpoints with the expression of PIMREG in BC were explored via TIMER, TISIDB, and GEPIA. Potential drugs that interact with PIMREG in BC were explored via Q-omic. The siRNA transfection, CCK-8, and transwell migration assay were conducted to explore the function of PIMREG in cell proliferation and migration. Results PIMREG expression was significantly higher in infiltrating ductal carcinoma, estrogen receptor negative BC, and progestin receptor negative BC. High expression of PIMREG was associated with poor overall survival, disease-specific survival, and progression-free interval. A nomogram based on PIMREG was developed with a satisfactory prognostic value. PIMREG also had a high diagnostic ability, with an area under the curve of 0.940. Its correlations with several immunomodulators were also observed. Immune checkpoint CTLA-4 was significantly positively associated with PIMREG. HDAC2 was found as a potentially critical link between PIMREG and BRCA1/2. In addition, PIMREG knockdown could inhibit cell proliferation and migration in BC. Conclusions The high expression of PIMREG is associated with poor prognosis and immune checkpoints in BC. HDAC2 may be a critical link between PIMREG and BRCA1/2, potentially a therapeutic target.
Collapse
Affiliation(s)
- Wenjing Zhao
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yuanjin Chang
- School of Medicine, Jiangnan College, WuXi, JiangSu, China
| | - Zhaoye Wu
- School of Medicine, Jiangnan College, WuXi, JiangSu, China
| | - Xiaofan Jiang
- School of Medicine, Jiangnan College, WuXi, JiangSu, China
| | - Yong Li
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ruijin Xie
- School of Medicine, Jiangnan College, WuXi, JiangSu, China
| | - Deyuan Fu
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Chenyu Sun
- Department of General Surgery, The second Affiliated Hospital of Anhui Medical University, Anhui, China
- Department of Medicine, AMITA Health Saint Joseph Hospital, Chicago, IL, USA
| | - Ju Gao
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
11
|
Moghtaderi M, Bazzazan S, Sorourian G, Sorourian M, Akhavanzanjani Y, Noorbazargan H, Ren Q. Encapsulation of Thymol in Gelatin Methacryloyl (GelMa)-Based Nanoniosome Enables Enhanced Antibiofilm Activity and Wound Healing. Pharmaceutics 2023; 15:1699. [PMID: 37376147 DOI: 10.3390/pharmaceutics15061699] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/31/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Non-healing wounds impose huge cost on patients, healthcare, and society, which are further fortified by biofilm formation and antimicrobial resistance (AMR) problems. Here, Thymol, an herbal antimicrobial agent, is utilized to combat AMR. For efficient delivery of Thymol gelatin methacryloyl (GelMa), a hydrophilic polymeric hydrogel with excellent biocompatibility combined with niosome was used to encapsulate Thymol. After optimization of the niosomal Thymol (Nio-Thymol) in the company of GelMa (Nio-Thymol@GelMa) to achieve maximum entrapment efficiency, minimum size, and low polydispersity index, the Thymol release peaked at 60% and 42% from Nio-Thymol@GelMa in medium with pH values of 6.5 and 7.4 after 72 h, respectively. Furthermore, Nio-Thymol@GelMa demonstrated higher antibacterial and anti-biofilm activity than Nio-Thymol and free Thymol against both Gram-negative and Gram-positive bacteria. Interestingly, compared with other obtained formulations, Nio-Thymol@GelMa also led to greater enhancement of migration of human dermal fibroblasts in vitro, and higher upregulation of the expression of certain growth factors such as FGF-1, and matrix metalloproteinases such as MMP-2 and MMP-13. These results suggest that Nio-Thymol@GelMa can represent a potential drug preparation for Thymol to enhance the wound healing process and antibacterial efficacy.
Collapse
Affiliation(s)
- Maryam Moghtaderi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 1417935840, Iran
| | - Saba Bazzazan
- Department of Community Medicine, Mashhad Branch, Islamic Azad University, Mashhad 1477893855, Iran
| | - Ghazal Sorourian
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 1417935840, Iran
| | - Maral Sorourian
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 1417935840, Iran
| | - Yasaman Akhavanzanjani
- Department of Molecular and Cellular Biology, Faculty of Advance Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran 1477893855, Iran
| | - Hassan Noorbazargan
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1517964311, Iran
| | - Qun Ren
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland
| |
Collapse
|
12
|
Venkatas J, Singh M. Curcumin-reduced gold nanoparticles facilitate IL-12 delivery to a cervical cancer in vitro cell model. Nanomedicine (Lond) 2023; 18:945-960. [PMID: 37503889 DOI: 10.2217/nnm-2023-0076] [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] [Indexed: 07/29/2023] Open
Abstract
Aim: To synthesize curcumin-reduced gold nanoparticles (AuNPs) for the efficient delivery to and expression of the IL-12 gene in cervical cancer (HeLa) cells in vitro. Methods: Curcumin-reduced AuNPs were synthesized, stabilized with poly-L-lysine and PEG, conjugated to IL-12 DNA and physicochemically characterized. Cytotoxicity and IL-12 expression were accessed in vitro. Results & discussion: Stable, spherical AuNPs effectively compacted and protected the IL-12 DNA and tolerated well in vitro. Real-time quantitative PCR and ELISA confirmed the successful delivery and expression of the IL-12 gene in HeLa cells. Conclusion: The favorable attributes of this AuNP-delivery system and the significant IL-12 expression obtained augur well for cytokine-based therapy or immunotherapy in cervical cancer.
Collapse
Affiliation(s)
- Jeaneen Venkatas
- Nano-Gene & Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, KwaZulu-Natal, 4000, South Africa
| | - Moganavelli Singh
- Nano-Gene & Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, KwaZulu-Natal, 4000, South Africa
| |
Collapse
|
13
|
Zani CP, Zani AP, Thomazini CM, Retamiro KM, de Oliveira AR, Gonçalves DL, Sarragiotto MH, Garcia FP, de Oliveira Silva S, Nakamura CV, Ueda-Nakamura T. β-Carboline-α-aminophosphonate Derivative: A Promising Antitumor Agent for Breast Cancer Treatment. Molecules 2023; 28:molecules28093949. [PMID: 37175359 PMCID: PMC10179861 DOI: 10.3390/molecules28093949] [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: 03/22/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Breast cancer is the most common type of cancer and the leading cause of cancer mortality among women worldwide. Considering the limitations of the current treatments available, we analyzed the in vitro cytotoxic potential of ((4-Fluoro-phenyl)-{2-[(1-phenyl-9H-β-carboline-3-carbonyl)-amino]-ethylamino}-methyl)-phosphonic acid dibutyl ester (BCP-1) in breast cancer cells (MCF-7 and MDA-MB-231) and in a non-tumor breast cell line (MCF-10A). BCP-1 has an α-aminophosphonate unit linked to the β-carboline nucleus, and the literature indicates that compounds of these classes have high biological potential. In the present study, the mechanism of action of BCP-1 was investigated through methods of spectrofluorimetry, flow cytometry, and protein expression analysis. It was found that BCP-1 inhibited the proliferation of both cancer cell lines. Furthermore, it induced oxidative stress and cell cycle arrest in G2/M. Upregulation of apoptosis-related proteins such as Bax, cytochrome C, and caspases, as well as a decrease in the anti-apoptotic protein Bcl-2, indicated potential induction of apoptosis in the MDA-MB-231 cells. While in MCF-7 cells, BCP-1 activated the autophagic death pathway, which was demonstrated by an increase in autophagic vacuoles and acidic organelles, in addition to increased expression of LC3I/LC3II and reduced SQSTM1/p62 expression. Further, BCP-1 demonstrated antimetastatic potential by reducing MMP-9 expression and cell migration in both breast cancer cell lines. In conclusion, BCP-1 is a promising candidate for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Caroline Pinto Zani
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Aline Pinto Zani
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Cristiane Melissa Thomazini
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Karina Miyuki Retamiro
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | | | - Débora Laís Gonçalves
- Department of Chemistry, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | | | - Francielle Pelegrin Garcia
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Sueli de Oliveira Silva
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Celso Vataru Nakamura
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Tania Ueda-Nakamura
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| |
Collapse
|
14
|
Rezaei N, Kazem Arki M, Miri-Lavasani Z, Solhi R, Khoramipour M, Rashedi H, Asadzadeh Aghdaei H, Hossein-Khannazer N, Mostafavi E, Vosough M. Co-delivery of Doxorubicin and Paclitaxel via Noisome Nanocarriers Attenuates Cancerous Phenotypes in Gastric Cancer Cells. Eur J Pharm Biopharm 2023:S0939-6411(23)00102-9. [PMID: 37105361 DOI: 10.1016/j.ejpb.2023.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Gastric cancer (GC) is known as a deadly malignancy all over the world, yet none of the current therapeutic regimens have achieved efficacy. this current study has aimed to optimize and reduce treatment doses and overcome multidrug resistance in GC by developing optimum niosomal formulation for the delivery of doxorubicin (DXR), paclitaxel (PTX), and their co-delivery. The particles' size, polydispersity index (PDI), and entrapment efficacy (EE%) were optimized using statistical techniques, i.e., Box-Behnken and Central Composite Design. In contrast to soluble drug formulations, the release rate of medicines from nanoparticles were higher in physiological and acidic pH. Niosomes were more stable at 4°C, compared to 25°C. The MTT assay revealed that the IC50 of drug-loaded niosomes was the lowest among all developed formulations. The apoptosis-related genes (CASPASE-3, CASPASE-8, and CASPASE-9) and tumor suppressor genes (BAX, BCL2) were evaluated in cancer cells before and after treatment. In comparison to control cells and cells treated with soluble forms of DXR and PTX, while the expression of BCL2 decreased, the expression of BAX, CASPASE-3, CASPASE-8, and CASPASE-9 was enhanced in cells treated with drug-loaded niosomes. Drug-loaded niosomes inhibited colony formation capacity and increased apoptosis in human AGS gastric cancer cells. Our results indicate that co-delivery of DXR and PTX-loaded niosomes may be an effective and innovative therapeutic approach to gastric cancer.
Collapse
Affiliation(s)
- Niloofar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mandana Kazem Arki
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohre Miri-Lavasani
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Solhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Khoramipour
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Rashedi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
15
|
Development and In Vitro and In Vivo Evaluation of an Antineoplastic Copper(II) Compound (Casiopeina III-ia) Loaded in Nonionic Vesicles Using Quality by Design. Int J Mol Sci 2022; 23:ijms232112756. [PMID: 36361549 PMCID: PMC9655312 DOI: 10.3390/ijms232112756] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 12/02/2022] Open
Abstract
In recent decades, the interest in metallodrugs as therapeutic agents has increased. Casiopeinas are copper-based compounds that have been evaluated in several tumor cell lines. Currently, casiopeina III-ia (CasIII-ia) is being evaluated in phase I clinical trials. The aim of the present work is to develop a niosome formulation containing CasIII-ia for intravenous administration through a quality-by-design (QbD) approach. Risk analysis was performed to identify the factors that may have an impact on CasIII-ia encapsulation. The developed nanoformulation optimized from the experimental design was characterized by spectroscopy, thermal analysis, and electronic microscopy. In vitro drug release showed a burst effect followed by a diffusion-dependent process. The niosomes showed physical stability for at least three months at 37 °C and 75% relative humidity. The in vitro test showed activity of the encapsulated CasIII-ia on a metastatic breast cancer cell line and the in vivo test of nanoencapsulated CasIII-ia maintained the activity of the free compound, but showed a diminished toxicity. Therefore, the optimal conditions obtained by QbD may improve the scaling-up process.
Collapse
|
16
|
Moghtaderi M, Sedaghatnia K, Bourbour M, Fatemizadeh M, Salehi Moghaddam Z, Hejabi F, Heidari F, Quazi S, Farasati Far B. Niosomes: a novel targeted drug delivery system for cancer. Med Oncol 2022; 39:240. [PMID: 36175809 DOI: 10.1007/s12032-022-01836-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/27/2022] [Indexed: 10/25/2022]
Abstract
Recently, nanotechnology is involved in various fields of science, of which medicine is one of the most obvious. The use of nanoparticles in the process of treating and diagnosing diseases has created a novel way of therapeutic strategies with effective mechanisms of action. Also, due to the remarkable progress of personalized medicine, the effort is to reduce the side effects of treatment paths as much as possible and to provide targeted treatments. Therefore, the targeted delivery of drugs is important in different diseases, especially in patients who receive combined drugs, because the delivery of different drug structures requires different systems so that there is no change in the drug and its effectiveness. Niosomes are polymeric nanoparticles that show favorable characteristics in drug delivery. In addition to biocompatibility and high absorption, these nanoparticles also provide the possibility of reducing the drug dosage and targeting the release of drugs, as well as the delivery of both hydrophilic and lipophilic drugs by Niosome vesicles. Since various factors such as components, preparation, and optimization methods are effective in the size and formation of niosomal structures, in this review, the characteristics related to niosome vesicles were first examined and then the in silico tools for designing, prediction, and optimization were explained. Finally, anticancer drugs delivered by niosomes were compared and discussed to be a suitable model for designing therapeutic strategies. In this research, it has been tried to examine all the aspects required for drug delivery engineering using niosomes and finally, by presenting clinical examples of the use of these nanocarriers in cancer, its clinical characteristics were also expressed.
Collapse
Affiliation(s)
- Maryam Moghtaderi
- Department of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Kamand Sedaghatnia
- Department of Applied Chemistry, Azad University of Tehran South Branch, Tehran, Iran
| | - Mahsa Bourbour
- Department of Biotechnology, Alzahra University, Tehran, Iran
| | - Mahdi Fatemizadeh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi Moghaddam
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Faranak Hejabi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Fatemeh Heidari
- Department of Cellular and Molecular Biology, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Sameer Quazi
- GenLab Biosolutions Private Limited, Bangalore, Karnataka, India.
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge, UK.
- Clinical Bioinformatics, School of Health Sciences, The University of Manchester, Manchester, UK.
| | - Bahareh Farasati Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| |
Collapse
|
17
|
Formulation and Characterization of Poly (Ethylene Glycol)-Coated Core-Shell Methionine Magnetic Nanoparticles as a Carrier for Naproxen Delivery: Growth Inhibition of Cancer Cells. Cancers (Basel) 2022; 14:cancers14071797. [PMID: 35406569 PMCID: PMC8997395 DOI: 10.3390/cancers14071797] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Naproxen was loaded onto a magnetic nanoparticle coated with polyethylene glycol. Magnetic nanoparticles (MNPs) were used in this study to develop a smart naproxen delivery system. One of the most potent COX-1 and COX-2 inhibitors is naproxen, which belongs to the NSAID family of drugs. Although this drug has a short half-life, it has considerable toxicities and side effects on gastrointestinal tissues. The significant potential of our proposed nanocarrier for biomedical applications has been widely recognized; we modified MNPs to attach to this drug via disulfide bonds, promote the selective release of naproxen in inflammatory cells, and prevent adverse effects on the digestive system. It was found that the cytotoxicity of the drug was lowered by this change, which prevented unspecific protein binding. Abstract An efficient and selective drug delivery vehicle for cancer cells can remarkably improve therapeutic approaches. In this study, we focused on the synthesis and characterization of magnetic Ni1−xCoxFe2O4 nanoparticles (NPs) coated with two layers of methionine and polyethylene glycol to increase the loading capacity and lower toxicity to serve as an efficient drug carrier. Ni1−xCoxFe2O4@Methionine@PEG NPs were synthesized by a reflux method then characterized by FTIR, XRD, FESEM, TEM, and VSM. Naproxen was used as a model drug and its loading and release in the vehicles were evaluated. The results for loading efficiency showed 1 mg of Ni1−xCoxFe2O4@Methionine@PEG NPs could load 0.51 mg of the naproxen. Interestingly, Ni1−xCoxFe2O4@Methionine@PEG showed a gradual release of the drug, achieving a time-release up to 5 days, and demonstrated that a pH 5 release of the drug was about 20% higher than Ni1−xCoxFe2O4@Methionine NPs, which could enhance the intracellular drug release following endocytosis. At pH 7.4, the release of the drug was slower than Ni1−xCoxFe2O4@Methionine NPs; demonstrating the potential to minimize the adverse effects of anticancer drugs on normal tissues. Moreover, naproxen loaded onto the Ni1−xCoxFe2O4@Methionine@PEG NPs for breast cancer cell lines MDA-MB-231 and MCF-7 showed more significant cell death than the free drug, which was measured by an MTT assay. When comparing both cancer cells, we demonstrated that naproxen loaded onto the Ni1−xCoxFe2O4@Methionine@PEG NPs exhibited greater cell death effects on the MCF-7 cells compared with the MDA-MB-231 cells. The results of the hemolysis test also showed good hemocompatibility. The results indicated that the prepared magnetic nanocarrier could be suitable for controlled anticancer drug delivery.
Collapse
|
18
|
Tang Y, Wang H, He Q, Chen Y, Wang J. Bioinformatics Method Was Used to Analyze the Highly Expressed Gene FAM83A of Breast Cancer in Young Women. Appl Bionics Biomech 2022; 2022:5358030. [PMID: 35392358 PMCID: PMC8983250 DOI: 10.1155/2022/5358030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 11/29/2022] Open
Abstract
Objectives Preliminary analysis of breast cancer related to unknown functional gene FAM83A through bioinformatics knowledge to inform further experimental studies. Select high expression genes for breast cancer and use bioinformatics methods to predict the biological function of FAM83A. Methods Genes with significant differences in expression between breast tumors and normal breast tissue libraries were selected from CGAP's SAGE Digital Gene Expression Displayer (DGED) database. An unknown functional gene, FAM83A, which is highly expressed in breast cancer, was screened. We performed an analysis of the gene structure, subcellular localization, physicochemical properties of the encoding products, functional sites, protein structure, and functional domains. Results Through SAGE DGED, a total of 185 genes with expression differences were found. The structure and function of FAM83A have ideal predictions, and it is generally determined that this gene encodes a nuclear protein with a nucleoprotein. The active site of PLDc and the functional domain of DUF1669 can be involved in signal transduction and gene expression regulation in tumorigenesis and metastasis. Digital gene representation of the Tumor Genome Project Data Library was used to select differentially expressed genes in breast cancer tissue and breast benign tumor tissue. Conclusion Studies show that FAM83A is a potential research target associated with tumorigenesis and metastasis. Initial tests confirmed the expression of this gene. Lay a solid foundation for further research learning. FAM83A is a highly expressed gene in breast cancer and can serve as a target for studying molecular mechanisms in breast cancer.
Collapse
Affiliation(s)
- Yongzhe Tang
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Hao Wang
- Teaching Center of Experimental Medicine, Shanghai Medical College, Fudan University, 138 Yixueyuan Rd, Shanghai 200032, China
| | - Qi He
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Yuanyuan Chen
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Jie Wang
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| |
Collapse
|