Sérgio Antunes Filho, Clara M Almeida, Maria Teresa Villela Romanos, Bianca Pizzorno Backx, Raquel Regina Bonelli
{"title":"Green synthesis of silver nanoparticles for functional cotton fabrics: antimicrobial efficacy against multidrug-resistant bacteria and cytotoxicity evaluation.","authors":"Sérgio Antunes Filho, Clara M Almeida, Maria Teresa Villela Romanos, Bianca Pizzorno Backx, Raquel Regina Bonelli","doi":"10.1080/21691401.2025.2485115","DOIUrl":"10.1080/21691401.2025.2485115","url":null,"abstract":"<p><p>Bacterial infections associated with healthcare are a challenge on a global scale due to the high morbidity and mortality rates, especially those caused by multidrug-resistant isolates. Hospital textiles are abiotic surfaces that may serve as a means of disseminating and persisting microorganisms in hospitals, as microorganisms can remain viable on these surfaces for up to months. In this study, we employed a green synthesis approach utilizing guava leaf extract (<i>Psidium guajava</i>) to produce silver nanoparticles, which were then incorporated into a cotton fabric. Antimicrobial properties and the cytotoxicity of the functional textile were assessed. The finding indicated that the green synthesis method was efficient and resulted in a predominant population of nanoparticles with diameters ranging from 25 to 84 nm that were uniformly dispersed in the textile. The functional textile exhibited low toxicity and high antimicrobial efficiency, even against multidrug-resistant microorganisms of particular concern in hospital settings. Atomic force microscopy carried out evidenced invaginations in the cell wall of bacteria submitted to this textile, suggesting surface damage as an important mechanism of action silver nanoparticles incorporated.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":"53 1","pages":"153-165"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143750924","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Maryam Fekri Soufiabadi, Reza Haji Hosseini, Zolfaghar Lotfi
{"title":"Investigating the anticancer and synergistic effects of the combination of green synthesized silver nanoparticles and papaverine on breast cancer cell lines: focusing on the apoptosis pathway and microRNA regulation.","authors":"Maryam Fekri Soufiabadi, Reza Haji Hosseini, Zolfaghar Lotfi","doi":"10.1080/21691401.2025.2507372","DOIUrl":"https://doi.org/10.1080/21691401.2025.2507372","url":null,"abstract":"<p><p>The study investigates the anticancer effects of green silver nanoparticles (Ag-NPs) synthesized from <i>Viola cornuta</i> extract combined with papaverine on breast cancer cells. Ag-NPs were characterized using various analytical techniques, confirming their presence with UV-vis spectroscopy showing a peak at 413 nm and an average size of 42 nm via field emission scanning electron microscopy (FE-SEM) analysis. The particles demonstrated a face-centred cubic structure, with energy-dispersive X-ray spectroscopy (EDX) confirming elemental composition. Additionally, the zeta potential measurement of -6.75 mV indicated favourable electrostatic repulsion between nanoparticles, thereby confirming their stability. Antioxidant activity was significant, with an EC<sub>50</sub> value of 38.78 μg/mL. The combination treatment of Ag-NPs and papaverine exhibited synergistic effects, lowering IC<sub>50</sub> values to 2.8 + 112.7 μg/mL for MCF-7 cells and 6.2 + 112 μg/mL for MDA-MB-231 cells, without toxicity to normal cells. Flow cytometry revealed G0/G1 phase inhibition and increased sub-G1 populations, indicating cell cycle arrest, alongside increased reactive oxygen species generation and apoptosis. Notably, the experimental group showed altered expression of oncogenic and tumour suppressor microRNAs and apoptotic genes (<i>p</i> < .0001), underscoring the potential of this nanoparticle-papaverine combination as an effective anticancer strategy against breast cancer treatment resistance.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":"53 1","pages":"1-19"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144198213","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"An integrated <i>in vitro</i> and <i>in silico</i> approach to assess targeted cytotoxicity against MDA-MB-231 triple-negative breast cancer cells with <i>Psidium guajava</i> peel-derived chitosan nanoparticles.","authors":"Vino Udappusamy, Rajan Thinagaran, Vijayakumar Mayakrishnan, Janani Balakarthikeyan, Priya Kannappan, Sameer Al-Ghamdi, Naif Abdurhman Alrudian, Mohammed Saad Alqahtani, Khalid Albasheer, Chandrabose Sureka, Mahmoud H El-Bidawy, Nesreen Alsanousi, Sahar Gamil, Thiyagarajan Ramesh","doi":"10.1080/21691401.2025.2462333","DOIUrl":"10.1080/21691401.2025.2462333","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is a significant global health issue, with high mortality rates. The chemotherapeutic drugs currently used for TNBC have significant side effects, impacting both normal and cancer cells. In this study, we investigated a potential use of fruit peel extract of <i>Psidium guajava</i> (PGP) encapsulated with chitosan nanoparticles (CSNPs) to combat TNBC. The synthesized PGP-CSNPs were characterized using UV-vis spectroscopy, Fourier transform infra-red (FTIR) spectroscopy, TEM and GC-MS. The maximum loading capacity and encapsulation efficacy of PGP-CSNPs were found to be 72.5 ± 0.49% and 92.9 ± 0.10%, respectively. Furthermore, <i>in vitro</i> cytotoxicity was assessed, and the IC<sub>50</sub> value for PGP-CSNPs was 50.13 µg/mL. It was observed that PGP-CSNPs could induce apoptosis in MDA-MB-231 cells in dose-dependent manner. Furthermore, molecular docking was performed for bioactive compounds retrieved from PGP-CSNPs against human tumour suppressor proteins Bcl2, and results showed that the PGP-CSNPs had lower binding energy than cisplatin. This suggests that, the synthesized PGP-CSNPs have the potential to serve as a therapeutic agent for tackling TNBC. However, to validate its efficacy in human therapy, furthermore pre-clinical and clinical procedures should be examined, as this is an ongoing and significant step towards developing an effective and safe anticancer drug.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":"53 1","pages":"43-55"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381561","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shiyao Liao, Kai Zhou, Yao Kang, Tingxiao Zhao, Yicheng Lin, Jun Lv, Danjie Zhu
{"title":"Enhanced cartilage repair using gelatin methacryloyl hydrogels combined with icariin and magnesium-doped bioactive glass.","authors":"Shiyao Liao, Kai Zhou, Yao Kang, Tingxiao Zhao, Yicheng Lin, Jun Lv, Danjie Zhu","doi":"10.1080/21691401.2025.2490677","DOIUrl":"https://doi.org/10.1080/21691401.2025.2490677","url":null,"abstract":"<p><p>Cartilage repair remains challenging due to limited self-healing, poor biocompatibility, and insufficient mechanical properties of current materials. To overcome these issues, we developed a multifunctional composite hydrogel by integrating gelatine methacrylate (GelMA) with magnesium-doped bioactive glass (Mg-BG) and icariin (ICA). SEM analysis revealed that pure GelMA exhibited a highly porous yet loosely organized structure, whereas the addition of Mg-BG and ICA produced a denser, more interconnected porous network that enhances cell adhesion and nutrient diffusion. <i>In vitro</i>, the ICA/Mg-BG/GelMA hydrogel achieved a swelling ratio up to 430% and maintained cell viability above 80% over 5 days. Moreover, qRT-PCR and immunohistochemical analyses demonstrated that the composite hydrogel upregulated chondrogenic markers (SOX9, ACAN, and COL2A1) compared with GelMA alone. Specifically, it downregulates M1 pro-inflammatory markers (CCR7, iNOS, CD86) and upregulates M2 anti-inflammatory markers (ARG1, CD163, CD206), thereby creating a regenerative microenvironment. These results indicate that the synergistic combination of GelMA, Mg-BG, and ICA not only improves the scaffold's mechanical support but also enhances its biological functionality, offering a promising strategy for cartilage repair. Future studies will focus on <i>in vivo</i> validation to further assess its clinical potential.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":"53 1","pages":"181-193"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143959826","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shela Salsabila, Miski Aghnia Khairinisa, Nasrul Wathoni, Irna Sufiawati, Wan Ezumi Mohd Fuad, Nur Kusaira Khairul Ikram, Muchtaridi Muchtaridi
{"title":"<i>In vivo</i> toxicity of chitosan-based nanoparticles: a systematic review.","authors":"Shela Salsabila, Miski Aghnia Khairinisa, Nasrul Wathoni, Irna Sufiawati, Wan Ezumi Mohd Fuad, Nur Kusaira Khairul Ikram, Muchtaridi Muchtaridi","doi":"10.1080/21691401.2025.2462328","DOIUrl":"10.1080/21691401.2025.2462328","url":null,"abstract":"<p><p>Chitosan nanoparticles have been extensively utilised as polymeric drug carriers in nanoparticles formulations due to their potential to enhance drug delivery, efficacy, and safety. Numerous toxicity studies have been previously conducted to assess the safety profile of chitosan-based nanoparticles. These toxicity studies employed various methodologies, including test animals, interventions, and different routes of administration. This review aims to summarise research on the safety profile of chitosan-based nanoparticles in drug delivery, with a focus on general toxicity tests to determine LD50 and NOAEL values. It can serve as a repository and reference for chitosan-based nanoparticles, facilitating future research and further development of drugs delivery system using chitosan nanoparticles. Publications from 2014 to 2024 were obtained from PubMed, Scopus, Google Scholar, and ScienceDirect, in accordance with the inclusion and exclusion criteria.The ARRIVE 2.0 guidelines were employed to evaluate the quality and risk-of-bias in the <i>in vivo</i> toxicity studies. The results demonstrated favourable toxicity profiles, often exhibiting reduced toxicity compared to free drugs or substances. Acute toxicity studies consistently reported high LD50 values, frequently exceeding 5000 mg/kg body weight, while subacute studies typically revealed no significant adverse effects. Various routes of administration varied, including oral, intravenous, intraperitoneal, inhalation, and topical, each demonstrating promising safety profiles.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":"53 1","pages":"1-15"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381557","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Prince N Dlozi, Rami Ahmed, Star Khoza, Admire Dube
{"title":"Vitamin D3 loaded polycaprolactone nanoparticles enhance the expression of the antimicrobial peptide cathelicidin in macrophages.","authors":"Prince N Dlozi, Rami Ahmed, Star Khoza, Admire Dube","doi":"10.1080/21691401.2025.2499515","DOIUrl":"https://doi.org/10.1080/21691401.2025.2499515","url":null,"abstract":"<p><p>Tuberculosis (TB), primarily caused by <i>Mycobacterium tuberculosis</i>, remains a global health burden. Current antibiotic treatments are limited by adverse effects, poor adherence, and drug resistance, necessitating new therapeutic approaches. Recent studies highlight the role of vitamin D3 (VD3) in enhancing host immune responses against the mycobacterium <i>via</i> cathelicidin (an antimicrobial peptide) and autophagy activation. In this study, VD3-loaded poly-ƹ-caprolactone (PCL) nanoparticles (NPs) were synthesized to enhance cathelicidin expression in macrophages. NPs containing cholecalciferol, calcifediol, and calcitriol were synthesized using an emulsification solvent-evaporation technique. Average sizes of synthesized NPs ranged from 304.7 to 458.7 nm, with polydispersity index (PDI) and zeta potential (ZP) ranging from 0.103 to 0.257 and -17.3 to -7.47 mV, respectively. Encapsulation efficiencies were 9.68%, 10.99%, and 19.28% for cholecalciferol, calcifediol, and calcitriol, respectively. VD3-encapsulated NPs stimulated a dose-dependent increase in cathelicidin expression in THP-1 macrophages. Encapsulated calcifediol and calcitriol (100 ng/ml) induced the expression of 243.46 ng/ml ± 4.55 ng/ml and 396.67 ng/ml ± 25.24 ng/ml of cathelicidin, respectively, which was significantly higher than that induced by the free drugs. These findings suggest that NP encapsulation may offer a more efficient approach to using vitamin D3 for inducing cathelicidin expression as a host-directed treatment for TB.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":"53 1","pages":"207-219"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12057764/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143966527","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Amira Mira, Tariq M Aljarba, Fatma M Abdel Bar, Rehab Ahmed, Walaa A Negm, Engy Elekhnawy, Hosam M El-Seadawy, Eman A Elmorsy, Salwa S Younis
{"title":"Antiparasitic and antimicrobial activity of <i>Ipomoea palmata</i> against <i>Toxoplasma gondii</i> and <i>Staphylococcus aureus</i>: correlation with major phenolics identified by HPLC.","authors":"Amira Mira, Tariq M Aljarba, Fatma M Abdel Bar, Rehab Ahmed, Walaa A Negm, Engy Elekhnawy, Hosam M El-Seadawy, Eman A Elmorsy, Salwa S Younis","doi":"10.1080/21691401.2025.2494796","DOIUrl":"https://doi.org/10.1080/21691401.2025.2494796","url":null,"abstract":"<p><p><i>Toxoplasma gondii</i>, a protozoan parasite found in water sources, causes toxoplasmosis, with no current protocols for inactivating its oocysts in water. <i>Staphylococcus aureus</i>, a significant bacterial pathogen, is known for causing various illnesses, including skin infections and biofilm-related diseases. This study investigated the antibacterial and antiparasitic properties of <i>Ipomoea palmata</i> leaf extract, rich in phenolics, against <i>T. gondii</i> tachyzoites and <i>S. aureus. I. palmata</i> extract significantly reduced tachyzoites count in peritoneal fluids and liver smears of infected mice with alleviation of toxoplasmosis-induced hepatitis. SEM showed surface irregularities in tachyzoites from treated groups. The extract demonstrated antibacterial action against <i>S. aureus</i> with a minimum inhibitory concentration of 128 to 512 <i>µ</i>g/mL, reduced biofilm formation from 69.23% to 15.38% of tested isolates, and downregulated biofilm genes (<i>cna</i>, <i>fnbA</i>, and <i>ica</i>) in 53.85% of isolates. Treatment with <i>I. palmata</i> extract improved liver architecture, reduced inflammation, and eliminated blood vessel congestion. The main phenolic acids identified by HPLC/UV analysis were chlorogenic acid, gallic acid, ellagic acid, and methyl gallate, while the predominant flavonoids were apigenin, quercetin, and naringenin. These findings highlight the potential of <i>I. palmata</i> extract as a natural antimicrobial and antiparasitic agent, warranting further research to isolate and evaluate its active compounds.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":"53 1","pages":"194-206"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143961925","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Machine learning-based integration develops a disulfidptosis-related lncRNA signature for improving outcomes in gastric cancer.","authors":"Tianze Zhang, Yuqing Chen, Zhiping Xiang","doi":"10.1080/21691401.2024.2440415","DOIUrl":"https://doi.org/10.1080/21691401.2024.2440415","url":null,"abstract":"<p><p>Gastric cancer remains one of the deadliest cancers globally due to delayed detection and limited treatment options, underscoring the critical need for innovative prognostic methods. Disulfidptosis, a recently discovered programmed cell death triggered by disulphide stress, presents a fresh avenue for therapeutic exploration. This research examines disulfidptosis-related long noncoding RNAs (DRLs) in gastric cancer, with the goal of leveraging these lncRNAs as potential markers to enhance patient outcomes and treatment approaches. Comprehensive genomic and clinical data from stomach adenocarcinoma (STAD) were obtained from The Cancer Genome Atlas (TCGA). Employing least absolute shrinkage and selection operator (LASSO) regression analysis, a prognostic model was devised incorporating five key DRLs to forecast survival rates. The effectiveness of this model was validated using Kaplan-Meier survival plots, receiver operating characteristic (ROC) curves, and extensive functional enrichment studies. The importance of select lncRNAs and the expression variability of genes tied to disulfidptosis were validated via quantitative real-time PCR (qRT-PCR) and Western blot tests, establishing a solid foundation for their prognostic utility. Analyses of functional enrichment and tumour mutation burden highlighted the biological importance of these DRLs, connecting them to critical cancer pathways and immune responses. These discoveries broaden our comprehension of the molecular framework of gastric cancer and bolster the development of tailored treatment plans, highlighting the substantial role of DRLs in clinical prognosis and therapeutic intervention.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":"53 1","pages":"1-13"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142863136","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jakub Jagielski, Karolina Dydak, Kaja Jaskot, Dmytro Soloviov, Maciej Kozak, Grzegorz Nowaczyk
{"title":"Antibacterial lipid liquid crystalline nanoparticles - synthesis and optimization by central composite design.","authors":"Jakub Jagielski, Karolina Dydak, Kaja Jaskot, Dmytro Soloviov, Maciej Kozak, Grzegorz Nowaczyk","doi":"10.1080/21691401.2025.2472928","DOIUrl":"10.1080/21691401.2025.2472928","url":null,"abstract":"<p><p>The rise of antibiotic-resistant bacteria demands new antimicrobial strategies. Glyceryl monolaurate (GML) shows antibacterial activity against Gram-positive bacteria like <i>S. aureus</i> but is ineffective against Gram-negative <i>E. coli</i> due to its outer membrane. GML's limited solubility and susceptibility to bacterial lipases hinder its direct use. This study developed glyceryl monooleate (GMO) lipid liquid crystalline nanoparticles (LLCNPs) incorporating GML to enhance its stability and efficacy. Using a central composite design (CCD), an optimal GMO:GML:F127 mass ratio of 26.5:3.5:1.5 was achieved. Characterization via dynamic light scattering (DLS), small angle X-ray scattering (SAXS), and cryo-transmission electron microscopy (cryo-TEM) confirmed the formation of bicontinuous cubic phase nanoparticles (<i>Pn3m</i> space group) with hydrophobic, hydrophilic, and amphiphilic regions, enabling the incorporation of diverse agents and the presence of sponge-like nanoparticles. The optimized LLCNPs inhibited <i>S. aureus</i> growth at concentrations ≥10 µg/mL by disrupting its membrane potential but showed no activity against <i>E. coli.</i> Cytotoxicity studies indicated that GML incorporation did not significantly affect cell viability compared to pure GMO LLCNPs. This nanoparticle system offers a biocompatible solution for treating Gram-positive bacterial infections and may synergize with existing antibiotics, warranting further investigation into its mechanisms and therapeutic potential.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":"53 1","pages":"69-86"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143603875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}