Pub Date : 2025-12-18DOI: 10.1016/j.colsurfb.2025.115386
Yuting Zhai , Jiaxiu Liu , Yangru Dai , Tianyang Li , Pingping Meng , Dongjiang Yang , Daohao Li , Shuchao Zhang
Traditional antibacterial strategies encounter challenges such as drug resistance and environmental pollution. In contrast, nanomaterial-based phototoxic therapies, including photodynamic therapy (PDT) and photothermal therapy (PTT), show promise in enhancing antibacterial efficacy and mitigating drug resistance. In this work, the potential and effectiveness of carbon quantum dot-modified molybdenum disulfide (CQDs@MoS2) composite has investigated for the rapid inactivation of pathogenic microorganisms in photodynamic and photothermal therapies. Under xenon lamp illumination, MoS2 generates reactive oxygen species (ROS) that damage bacterial membranes and DNA. The incorporation of CQDs generates localized heat under infrared light, enhances the charge separation of the composite, and increases ROS production, thus producing a PTT effect for amplifying photocatalytic and antibacterial activities. This approach significantly inactivates Gram positive Staphylococcus aureus (S.aureus) and Gram negative Escherichia coli (E. coli) within 5 min and demonstrates high stability. The synergistic effects of these therapies promote effective bacterial inactivation, presenting a promising solution for water disinfection and biomedical applications.
{"title":"Carbon quantum dot modified MoS2 mediating photodynamic and photothermal therapy in rapid inactivation of pathogenic microorganisms","authors":"Yuting Zhai , Jiaxiu Liu , Yangru Dai , Tianyang Li , Pingping Meng , Dongjiang Yang , Daohao Li , Shuchao Zhang","doi":"10.1016/j.colsurfb.2025.115386","DOIUrl":"10.1016/j.colsurfb.2025.115386","url":null,"abstract":"<div><div>Traditional antibacterial strategies encounter challenges such as drug resistance and environmental pollution. In contrast, nanomaterial-based phototoxic therapies, including photodynamic therapy (PDT) and photothermal therapy (PTT), show promise in enhancing antibacterial efficacy and mitigating drug resistance. In this work, the potential and effectiveness of carbon quantum dot-modified molybdenum disulfide (CQDs@MoS<sub>2</sub>) composite has investigated for the rapid inactivation of pathogenic microorganisms in photodynamic and photothermal therapies. Under xenon lamp illumination, MoS<sub>2</sub> generates reactive oxygen species (ROS) that damage bacterial membranes and DNA. The incorporation of CQDs generates localized heat under infrared light, enhances the charge separation of the composite, and increases ROS production, thus producing a PTT effect for amplifying photocatalytic and antibacterial activities. This approach significantly inactivates Gram positive Staphylococcus aureus (<em>S.aureus</em>) and Gram negative Escherichia coli (<em>E. coli</em>) within 5 min and demonstrates high stability. The synergistic effects of these therapies promote effective bacterial inactivation, presenting a promising solution for water disinfection and biomedical applications.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115386"},"PeriodicalIF":5.6,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145831848","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-18DOI: 10.1016/j.colsurfb.2025.115385
Isabella Walker, Huiyan Li
Early detection of diseases significantly improves patient outcomes, yet many biomarkers remain at ultra-low concentrations in body fluids during the initial stages, posing challenges for conventional diagnostic techniques. Extracellular vesicles (EVs) have emerged as promising non-invasive biomarkers due to their presence in body fluids such as blood, saliva and urine, and because their molecular cargo reflects their cells of origin. However, sensitive and accurate detection of disease related EVs is challenging because of their low concentrations and nanoscale size. Metal enhanced fluorescence (MEF) has recently been applied to overcome these limitations by amplifying the fluorescent signal of labeled EVs through localized surface plasmon resonances of metallic nanostructures. This amplification enables the detection and quantification of EV-associated biomarkers with improved sensitivity, offering potential for earlier disease diagnosis. However, to date, few reviews have focused specifically on MEF-based applications for the detection and quantification of EVs and their molecular cargo. This review examines recent advances in MEF-based platforms for EV analysis, including nanohole arrays, metal nano islands and nanoarrays, gold nanoparticles in three-dimensional matrices, metal-organic frameworks and other plasmonic approaches. While current reviews have discussed the use of MEF biosensing application, this article specifically focuses on MEF- based EV detection highlighting the design principles, optical properties, advantages, and limitations of these platforms. Collectively, MEF offers a powerful strategy to enhance EV detection, bridging the gap between experimental analysis and clinical application.
{"title":"Applications of metal enhanced fluorescence for the detection and quantification of extracellular vesicles","authors":"Isabella Walker, Huiyan Li","doi":"10.1016/j.colsurfb.2025.115385","DOIUrl":"10.1016/j.colsurfb.2025.115385","url":null,"abstract":"<div><div>Early detection of diseases significantly improves patient outcomes, yet many biomarkers remain at ultra-low concentrations in body fluids during the initial stages, posing challenges for conventional diagnostic techniques. Extracellular vesicles (EVs) have emerged as promising non-invasive biomarkers due to their presence in body fluids such as blood, saliva and urine, and because their molecular cargo reflects their cells of origin. However, sensitive and accurate detection of disease related EVs is challenging because of their low concentrations and nanoscale size. Metal enhanced fluorescence (MEF) has recently been applied to overcome these limitations by amplifying the fluorescent signal of labeled EVs through localized surface plasmon resonances of metallic nanostructures. This amplification enables the detection and quantification of EV-associated biomarkers with improved sensitivity, offering potential for earlier disease diagnosis. However, to date, few reviews have focused specifically on MEF-based applications for the detection and quantification of EVs and their molecular cargo. This review examines recent advances in MEF-based platforms for EV analysis, including nanohole arrays, metal nano islands and nanoarrays, gold nanoparticles in three-dimensional matrices, metal-organic frameworks and other plasmonic approaches. While current reviews have discussed the use of MEF biosensing application, this article specifically focuses on MEF- based EV detection highlighting the design principles, optical properties, advantages, and limitations of these platforms. Collectively, MEF offers a powerful strategy to enhance EV detection, bridging the gap between experimental analysis and clinical application.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115385"},"PeriodicalIF":5.6,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145799421","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1016/j.colsurfb.2025.115382
Hao Shen , Tong Li , Xueying Chen , Zhurun Fang , Yan Xu , Kai Zheng , Ming Zhang
As the primary physical and immunological barrier against external insults, skin integrity is frequently compromised by trauma, disease, or iatrogenic injury, leading to pathogenic microorganism invasion and infected wounds. The escalating challenges posed by bacterial antibiotic resistance and biofilm formation highlight the urgent need for therapeutic systems that synergistically combine potent antibacterial efficacy with effective tissue regeneration modulation. Therefore, this study developed a multifunctional hydrogel platform integrating rapid photothermal sterilization and pro-regenerative bioactivity. We engineered an aggregation-induced emission (AIE) nanoparticle-loaded type I recombinant humanized collagen hydrogel (AIE@RHCI), achieving rapid UV-triggered crosslinking from liquid to gel state. The hydrogel demonstrates appropriate swelling capacity, controllable degradation kinetics, and favorable mechanical properties, enabling stable adhesion to dynamically deforming skin during movement. The incorporated AIE component exhibits 39.7 % photothermal conversion efficiency under 808 nm near-infrared (NIR) irradiation. In vitro studies confirmed that the AIE@RHCI hydrogel under NIR irradiation reduced methicillin-resistant Staphylococcus aureus (MRSA) viability to 22.2 %, while concurrently achieving 79.8 % biofilm inhibition. Simultaneously, the recombinant collagen matrix promotes vascular endothelial cell migration and accelerates vascularization, with significant upregulation of key pro-angiogenic genes VEGF and CD31. This integrated platform synergistically combines photothermal bactericidal nanoparticles with bioactive recombinant collagen scaffolding, offering a novel and promising therapeutic strategy for the management of antibiotic-resistant wound infections, with potential for clinical translation in chronic and infected wound care.
{"title":"Multifunctional collagen hydrogel accelerates infected wound repair through photothermal disinfection and pro-angiogenic activity","authors":"Hao Shen , Tong Li , Xueying Chen , Zhurun Fang , Yan Xu , Kai Zheng , Ming Zhang","doi":"10.1016/j.colsurfb.2025.115382","DOIUrl":"10.1016/j.colsurfb.2025.115382","url":null,"abstract":"<div><div>As the primary physical and immunological barrier against external insults, skin integrity is frequently compromised by trauma, disease, or iatrogenic injury, leading to pathogenic microorganism invasion and infected wounds. The escalating challenges posed by bacterial antibiotic resistance and biofilm formation highlight the urgent need for therapeutic systems that synergistically combine potent antibacterial efficacy with effective tissue regeneration modulation. Therefore, this study developed a multifunctional hydrogel platform integrating rapid photothermal sterilization and pro-regenerative bioactivity. We engineered an aggregation-induced emission (AIE) nanoparticle-loaded type I recombinant humanized collagen hydrogel (AIE@RHCI), achieving rapid UV-triggered crosslinking from liquid to gel state. The hydrogel demonstrates appropriate swelling capacity, controllable degradation kinetics, and favorable mechanical properties, enabling stable adhesion to dynamically deforming skin during movement. The incorporated AIE component exhibits 39.7 % photothermal conversion efficiency under 808 nm near-infrared (NIR) irradiation. <em>In vitro</em> studies confirmed that the AIE@RHCI hydrogel under NIR irradiation reduced methicillin-resistant <em>Staphylococcus aureus</em> (MRSA) viability to 22.2 %, while concurrently achieving 79.8 % biofilm inhibition. Simultaneously, the recombinant collagen matrix promotes vascular endothelial cell migration and accelerates vascularization, with significant upregulation of key pro-angiogenic genes <em>VEGF</em> and <em>CD31</em>. This integrated platform synergistically combines photothermal bactericidal nanoparticles with bioactive recombinant collagen scaffolding, offering a novel and promising therapeutic strategy for the management of antibiotic-resistant wound infections, with potential for clinical translation in chronic and infected wound care.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115382"},"PeriodicalIF":5.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145792787","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1016/j.colsurfb.2025.115381
Yunong Shi , Miaosheng Tian , Shuo Shi , Jihua Gao
Inflammatory bowel disease (IBD) remains a therapeutic challenge due to its complex pathophysiology involving oxidative stress and chronic inflammation. Here, we developed a multifunctional hydrogel system (CeO2@Rh-gel) integrating rhein, a natural anti-inflammatory agent, and CeO2 NPs with ROS scavenging nanozyme activity for controlled release IBD therapy. The CeO2@Rh-gel was fabricated through self-assembly, forming a pH-responsive network that protected payloads in the stomach while enabling controlled release in the inflamed colon. In vitro, CeO2@Rh-gel exhibited synergistic antioxidant effects, mimicking SOD and CAT to eliminate ROS, while suppressing pro-inflammatory cytokines in LPS-stimulated macrophages. In a DSS-induced murine colitis model, oral CeO2@Rh-gel significantly alleviated disease severity, as evidenced by reduced weight loss, normalized colon length, and improved histopathology. Mechanistically, the hydrogel restored redox homeostasis and attenuated systemic inflammation. By combining ROS scavenging, anti-inflammatory action, and controllable delivery, CeO2@Rh-gel offers a promising dual-functional strategy for precision IBD therapy.
{"title":"pH-responsive phytochemical hydrogel loaded with antioxidant nanozymes for synergistic ROS scavenging and targeted inflammatory bowel disease therapy","authors":"Yunong Shi , Miaosheng Tian , Shuo Shi , Jihua Gao","doi":"10.1016/j.colsurfb.2025.115381","DOIUrl":"10.1016/j.colsurfb.2025.115381","url":null,"abstract":"<div><div>Inflammatory bowel disease (IBD) remains a therapeutic challenge due to its complex pathophysiology involving oxidative stress and chronic inflammation. Here, we developed a multifunctional hydrogel system (CeO<sub>2</sub>@Rh-gel) integrating rhein, a natural anti-inflammatory agent, and CeO<sub>2</sub> NPs with ROS scavenging nanozyme activity for controlled release IBD therapy. The CeO<sub>2</sub>@Rh-gel was fabricated through self-assembly, forming a pH-responsive network that protected payloads in the stomach while enabling controlled release in the inflamed colon. In vitro, CeO<sub>2</sub>@Rh-gel exhibited synergistic antioxidant effects, mimicking SOD and CAT to eliminate ROS, while suppressing pro-inflammatory cytokines in LPS-stimulated macrophages. In a DSS-induced murine colitis model, oral CeO<sub>2</sub>@Rh-gel significantly alleviated disease severity, as evidenced by reduced weight loss, normalized colon length, and improved histopathology. Mechanistically, the hydrogel restored redox homeostasis and attenuated systemic inflammation. By combining ROS scavenging, anti-inflammatory action, and controllable delivery, CeO<sub>2</sub>@Rh-gel offers a promising dual-functional strategy for precision IBD therapy.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115381"},"PeriodicalIF":5.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145814915","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1016/j.colsurfb.2025.115380
Su-Min Lee , Yunyoung Cho , Jiwoo Lim , Seungwoo Chung , Ngoc-Tu Nguyen , Yang-Hoon Kim , Sang-Ho Park , Jiho Min
Yeast vacuoles have recently emerged as promising bio-nanomaterials for drug delivery, offering improved stability and efficacy compared with traditional synthetic systems. Their membranes share structural and compositional similarities with those of mammalian cells, offering excellent biocompatibility and potential for efficient cellular interaction. These unique properties make reassembled vacuoles (ReV) attractive candidates for developing safe and effective therapeutic delivery platforms. This study optimized the vacuole reassembly process to enhance the performance of drug delivery. We compared two methods: Method 1, using conventional long-duration sonication and filtration, and Method 2, featuring an optimized 5-minute sonication without filtration. Reassembled vacuoles produced by method 1 (ReVMtd1), induced moderate TLR2 expression, suggesting mild immune priming without significant activation of inflammatory cytokines. In contrast, Reassembled vacuoles produced by method 2 (ReVMtd2) demonstrated superior immune activation, showing a dose-dependent upregulation of iNOS and TLR2. Additionally, ReVMtd2 achieved an encapsulation efficiency of 12.9 % for daunorubicin (DNR), comparable to native vacuoles, and maintained structural stability over a 12-month period. These findings highlight the potential of ReVMtd2 as a robust, biocompatible, and efficient drug delivery system, offering enhanced therapeutic performance and long-term stability.
{"title":"Fabrication of immune-enhancing vesicles from reassembled yeast vacuolar membranes","authors":"Su-Min Lee , Yunyoung Cho , Jiwoo Lim , Seungwoo Chung , Ngoc-Tu Nguyen , Yang-Hoon Kim , Sang-Ho Park , Jiho Min","doi":"10.1016/j.colsurfb.2025.115380","DOIUrl":"10.1016/j.colsurfb.2025.115380","url":null,"abstract":"<div><div>Yeast vacuoles have recently emerged as promising bio-nanomaterials for drug delivery, offering improved stability and efficacy compared with traditional synthetic systems. Their membranes share structural and compositional similarities with those of mammalian cells, offering excellent biocompatibility and potential for efficient cellular interaction. These unique properties make reassembled vacuoles (ReV) attractive candidates for developing safe and effective therapeutic delivery platforms. This study optimized the vacuole reassembly process to enhance the performance of drug delivery. We compared two methods: Method 1, using conventional long-duration sonication and filtration, and Method 2, featuring an optimized 5-minute sonication without filtration. Reassembled vacuoles produced by method 1 (ReV<sub>Mtd1</sub>), induced moderate TLR2 expression, suggesting mild immune priming without significant activation of inflammatory cytokines. In contrast, Reassembled vacuoles produced by method 2 (ReV<sub>Mtd2</sub>) demonstrated superior immune activation, showing a dose-dependent upregulation of iNOS and TLR2. Additionally, ReV<sub>Mtd2</sub> achieved an encapsulation efficiency of 12.9 % for daunorubicin (DNR), comparable to native vacuoles, and maintained structural stability over a 12-month period. These findings highlight the potential of ReV<sub>Mtd2</sub> as a robust, biocompatible, and efficient drug delivery system, offering enhanced therapeutic performance and long-term stability.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115380"},"PeriodicalIF":5.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145799423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Traumatic massive hemorrhage is a critical problem in the global emergency medicine field. Traditional hemostatic materials such as gauze and sponge have problems such as poor adhesion to irregular wounds and low adhesion strength. Existing hemostatic hydrogels also face bottlenecks such as insufficient active hemostatic efficiency, challenges in balancing interface adhesion and flexibility, and a lack of functional synergy, making it difficult to meet clinical needs. To address these challenges, this study develops a self-healing hydrogel spray that can form in situ for efficient hemostasis by simply mixing oxidized dextran (ODex) with polyethyleneimine (PEI) through a dynamic Schiff base reaction. Benefiting from the 'aldehyde-amino' active crosslinking mechanism, this hydrogel spray exhibits rapid gelation, triggering crosslinking within 15 s, with an adhesion strength exceeding 10 kPa, while also possessing good self-healing properties to adapt to dynamic and irregular wound. The electrostatic physical antibacterial properties of polyethyleneimine effectively suppress both Gram-positive and Gram-negative bacteria, avoiding the risk of drug resistance. By adjusting the oxidation degree of dextran, the hemostatic-adhesion synergy of the material has been further optimized, and the hydrogel preparation process is simple, cost-effective, user-friendly and can be stored at room temperature, with good biocompatibility. Animal experiments indicate that this hydrogel spray can quickly cover and adhere to the wound, effectively stopping bleeding, while reducing the risk of infection and promoting wound healing, providing innovative strategies and technical support for the development of a new generation of clinically available traumatic hemostatic materials.
{"title":"A user-friendly multifunctional hydrogel spray with adjustable mechanical properties for hemostasis and infected wound healing","authors":"Mengyao Gao , Zongliang Wang , Zhaohui Tang , Peibiao Zhang","doi":"10.1016/j.colsurfb.2025.115378","DOIUrl":"10.1016/j.colsurfb.2025.115378","url":null,"abstract":"<div><div>Traumatic massive hemorrhage is a critical problem in the global emergency medicine field. Traditional hemostatic materials such as gauze and sponge have problems such as poor adhesion to irregular wounds and low adhesion strength. Existing hemostatic hydrogels also face bottlenecks such as insufficient active hemostatic efficiency, challenges in balancing interface adhesion and flexibility, and a lack of functional synergy, making it difficult to meet clinical needs. To address these challenges, this study develops a self-healing hydrogel spray that can form in situ for efficient hemostasis by simply mixing oxidized dextran (ODex) with polyethyleneimine (PEI) through a dynamic Schiff base reaction. Benefiting from the 'aldehyde-amino' active crosslinking mechanism, this hydrogel spray exhibits rapid gelation, triggering crosslinking within 15 s, with an adhesion strength exceeding 10 kPa, while also possessing good self-healing properties to adapt to dynamic and irregular wound. The electrostatic physical antibacterial properties of polyethyleneimine effectively suppress both Gram-positive and Gram-negative bacteria, avoiding the risk of drug resistance. By adjusting the oxidation degree of dextran, the hemostatic-adhesion synergy of the material has been further optimized, and the hydrogel preparation process is simple, cost-effective, user-friendly and can be stored at room temperature, with good biocompatibility. Animal experiments indicate that this hydrogel spray can quickly cover and adhere to the wound, effectively stopping bleeding, while reducing the risk of infection and promoting wound healing, providing innovative strategies and technical support for the development of a new generation of clinically available traumatic hemostatic materials.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115378"},"PeriodicalIF":5.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145825382","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-16DOI: 10.1016/j.colsurfb.2025.115374
Chen-yan Li , Shuohua Xie , Min Deng , Ganrong Huang , Yanqiang Huang , Shufang Li
Objectives
Oral squamous cell carcinoma (OSCC) is a major malignancy affecting the oral, jaw, and facial regions. In this study, we synthesized a cinnamaldehyde-thiosemicarbazone-zinc (II) complex (CTZn) to inhibit OSCC cell proliferation.
Methods
We investigated the inhibitory effects of CTZn on OSCC using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, cell colony formation assays, wound-healing/scratch assays and transwell invasion assays. Flow cytometry (FCM) was performed to explore the CTZn's impact on apoptosis and cell-cycle progression. Through in vivo experiments, its antitumor activity activity was evaluated in OSCC xenograft mouse models and orthotopic tumor model mice. The mechanisms of CTZn were explored by detecting targeted metabolites, and performing drug affinity responsive target stability (DARTS) experiments, molecular docking analyses and Western blot assays. Levels of reactive oxygen species (ROS), oxidized and reduced nicotinamide adenine dinucleotide phosphate (NADP⁺/NADPH), glutathione (GSH), and 6-phosphogluconate dehydrogenase (PGD) protein in CAL-27 cells were also determined.
Results
These findings demonstrated that CTZn inhibited CAL-27 cell growth in vitro in a time- and concentration-dependent manner, with an IC50 value of 1.642–2.223μmol/L, and CTZn also exerted anti-tumor activity in vivo. CTZn also inhibited PGD messenger RNA (mRNA), and protein expression, reduced NADP+ /NADPH and GSH levels, and significantly increased ROS levels, thereby inducing oxidative stress.
Conclusion
CTZn impairs mitochondrial function, decreases ATP, levels, and induces G₂-phase arrest and apoptosis in CAL-27 cells. Therefore, it is an ideal drug for treating OSCC.
{"title":"The cinnamaldehyde-thiosemicarbazone-zinc (II) complex induces apoptosis in CAL-27 cells","authors":"Chen-yan Li , Shuohua Xie , Min Deng , Ganrong Huang , Yanqiang Huang , Shufang Li","doi":"10.1016/j.colsurfb.2025.115374","DOIUrl":"10.1016/j.colsurfb.2025.115374","url":null,"abstract":"<div><h3>Objectives</h3><div>Oral squamous cell carcinoma (OSCC) is a major malignancy affecting the oral, jaw, and facial regions. In this study, we synthesized a cinnamaldehyde-thiosemicarbazone-zinc (II) complex (CTZn) to inhibit OSCC cell proliferation.</div></div><div><h3>Methods</h3><div>We investigated the inhibitory effects of CTZn on OSCC using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, cell colony formation assays, wound-healing/scratch assays and transwell invasion assays. Flow cytometry (FCM) was performed to explore the CTZn's impact on apoptosis and cell-cycle progression. Through <em>in vivo</em> experiments, its antitumor activity activity was evaluated in OSCC xenograft mouse models and orthotopic tumor model mice. The mechanisms of CTZn were explored by detecting targeted metabolites, and performing drug affinity responsive target stability (DARTS) experiments, molecular docking analyses and Western blot assays. Levels of reactive oxygen species (ROS), oxidized and reduced nicotinamide adenine dinucleotide phosphate (NADP⁺/NADPH), glutathione (GSH), and 6-phosphogluconate dehydrogenase (PGD) protein in CAL-27 cells were also determined.</div></div><div><h3>Results</h3><div>These findings demonstrated that CTZn inhibited CAL-27 cell growth <em>in vitro</em> in a time- and concentration-dependent manner, with an IC<sub>50</sub> value of 1.642–2.223μmol/L, and CTZn also exerted anti-tumor activity <em>in vivo</em>. CTZn also inhibited PGD messenger RNA (mRNA), and protein expression, reduced NADP+ /NADPH and GSH levels, and significantly increased ROS levels, thereby inducing oxidative stress.</div></div><div><h3>Conclusion</h3><div>CTZn impairs mitochondrial function, decreases ATP, levels, and induces G₂-phase arrest and apoptosis in CAL-27 cells. Therefore, it is an ideal drug for treating OSCC.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115374"},"PeriodicalIF":5.6,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145799403","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Breast cancer continues to be the most common malignancy among women worldwide, requiring novel therapeutic approaches. This research investigates an innovative gene delivery strategy employing mesoporous silica nanoparticles (MCM-41) modified with lysine and cysteine (Lys-Cys) for the effective delivery of CRISPR-Cas9 plasmids aimed at the monocyte chemoattractant protein-1 (MCP-1/CCL2) gene. Bioinformatics analysis of the TCGA-BRCA dataset revealed substantial deregulation of CCL2 in breast cancer, underscoring its involvement in tumor growth and inflammation. The MCM/Lys-Cys nanocarrier demonstrated remarkable biocompatibility and effectively encapsulated a plasmid containing GFP, promoting superior cellular uptake in MDA-MB-231 breast cancer cells compared to conventional techniques. Functional experiments demonstrated that CRISPR/Cas9-mediated suppression of CCL2 markedly decreased cell proliferation, migration, and invasion, highlighting the promise of this targeted gene therapy strategy in breast cancer management. The findings indicate that the MCM/Lys-Cys nanosystem presents a viable non-viral approach for precise gene editing, potentially boosting therapeutic efforts against breast cancer by modulating inflammatory pathways.
{"title":"The MCM/Lys-Cys nanodevices for the efficient gene delivery: An approach towards MCP1 gene manipulation using CRISPR technology","authors":"Azadeh Rahimi , Ilnaz Rahimmanesh , Navid Abedpoor , Maryam Boshtam , Elham Bidram , Shaghayegh Haghjooy Javanmard , Hossein Khanahmad , Laleh Rafiee , Ashkan Bigham , Mohammad Rafienia , Saeed Karbasi , Laleh Shariati","doi":"10.1016/j.colsurfb.2025.115377","DOIUrl":"10.1016/j.colsurfb.2025.115377","url":null,"abstract":"<div><div>Breast cancer continues to be the most common malignancy among women worldwide, requiring novel therapeutic approaches. This research investigates an innovative gene delivery strategy employing mesoporous silica nanoparticles (MCM-41) modified with lysine and cysteine (Lys-Cys) for the effective delivery of CRISPR-Cas9 plasmids aimed at the monocyte chemoattractant protein-1 (MCP-1/CCL2) gene. Bioinformatics analysis of the TCGA-BRCA dataset revealed substantial deregulation of CCL2 in breast cancer, underscoring its involvement in tumor growth and inflammation. The MCM/Lys-Cys nanocarrier demonstrated remarkable biocompatibility and effectively encapsulated a plasmid containing GFP, promoting superior cellular uptake in MDA-MB-231 breast cancer cells compared to conventional techniques. Functional experiments demonstrated that CRISPR/Cas9-mediated suppression of CCL2 markedly decreased cell proliferation, migration, and invasion, highlighting the promise of this targeted gene therapy strategy in breast cancer management. The findings indicate that the MCM/Lys-Cys nanosystem presents a viable non-viral approach for precise gene editing, potentially boosting therapeutic efforts against breast cancer by modulating inflammatory pathways.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115377"},"PeriodicalIF":5.6,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145825404","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-15DOI: 10.1016/j.colsurfb.2025.115375
Siyu Du , Yi Zheng , Yabing Chen , Yijing Guo , Xinyue Kang , Lei Wang
The growing incidence of periodontitis demands innovative therapies, as current drug treatments are limited by short retention times, rapid clearance, and the risk of antimicrobial resistance. This study introduces a novel pH-responsive nano-miclle system for simvastatin (SIM) delivery. Its core innovation lies in being the first system ((SIM NP)n) of its kind to be stabilized by a cross-linked "outer shell," which significantly enhances its stability and controlled release capabilities. This "smart" system remains stable in a neutral environment (pH 7.4), preventing premature drug release. However, upon encountering the acidic inflammatory microenvironment of periodontitis, the shell degrades, triggering the sustained release of SIM directly at the site of inflammation. In vitro studies demonstrated that the SIM-loaded micelles possess potent immunomodulatory effects: they effectively suppressed pro-inflammatory M1 macrophage polarization (decreasing IL-1β, iNOS) while promoting anti-inflammatory M2 macrophage polarization (increasing Arg-1, IL-10). Mechanistic investigation confirmed this therapeutic effect is mediated by the PI3K/AKT/mTOR signaling pathway. More importantly, in a mouse model of periodontitis, the nano-micelles significantly reduced alveolar bone resorption, demonstrating potent anti-inflammatory and bone-protective efficacy in vivo. In conclusion, this "smart" pH-triggered delivery system for SIM offers a highly promising and effective strategy to overcome the limitations of traditional therapies, providing a new targeted path for the treatment of periodontitis.
{"title":"Fighting periodontitis with a pH-triggered nanocoating: A sustained-release strategy for simvastatin delivery","authors":"Siyu Du , Yi Zheng , Yabing Chen , Yijing Guo , Xinyue Kang , Lei Wang","doi":"10.1016/j.colsurfb.2025.115375","DOIUrl":"10.1016/j.colsurfb.2025.115375","url":null,"abstract":"<div><div>The growing incidence of periodontitis demands innovative therapies, as current drug treatments are limited by short retention times, rapid clearance, and the risk of antimicrobial resistance. This study introduces a novel pH-responsive nano-miclle system for simvastatin (SIM) delivery. Its core innovation lies in being the first system ((SIM NP)n) of its kind to be stabilized by a cross-linked \"outer shell,\" which significantly enhances its stability and controlled release capabilities. This \"smart\" system remains stable in a neutral environment (pH 7.4), preventing premature drug release. However, upon encountering the acidic inflammatory microenvironment of periodontitis, the shell degrades, triggering the sustained release of SIM directly at the site of inflammation. In vitro studies demonstrated that the SIM-loaded micelles possess potent immunomodulatory effects: they effectively suppressed pro-inflammatory M1 macrophage polarization (decreasing IL-1β, iNOS) while promoting anti-inflammatory M2 macrophage polarization (increasing Arg-1, IL-10). Mechanistic investigation confirmed this therapeutic effect is mediated by the PI3K/AKT/mTOR signaling pathway. More importantly, in a mouse model of periodontitis, the nano-micelles significantly reduced alveolar bone resorption, demonstrating potent anti-inflammatory and bone-protective efficacy in vivo. In conclusion, this \"smart\" pH-triggered delivery system for SIM offers a highly promising and effective strategy to overcome the limitations of traditional therapies, providing a new targeted path for the treatment of periodontitis.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115375"},"PeriodicalIF":5.6,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145792768","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-15DOI: 10.1016/j.colsurfb.2025.115373
Haoyang Gong , Xiaonan Li , Xinru Feng , Dongjing Wang , Xinyi Zhang , Kai Wang , Yanchen Liu , Lu Xu , Xueyan Zhou , Yanzhuo Zhang
Ultra-small copper sulfide (CuS) nanoparticles (NPs) possess exceptional photothermal performance, Fenton-like reaction activities, and multimodal imaging characteristics, while sodium nitroprusside serves as a clinical nitric oxide (NO) donor. A judicious combination of these two agents may facilitate the development of a novel multimodal synergistic antitumor strategy. In this context, we developed macrophage membrane-camouflaged and folic acid-conjugated nanocomposites (MF-SPC) that demonstrate dual responsiveness to near-infrared (NIR) laser and pH, aiming to enhance multimodal synergistic antitumor therapy. Within these biomimetic nanocomposites, sodium nitroprusside-doped Prussian blue NPs serve as the cores for inducing photothermal-responsive NO donation, while polydopamine layer embedded with CuS dots (sub-5 nm) act as the shells. This configuration not only enhances photothermal effects but also promotes Fenton-like reaction activity, glutathione depletion capabilities, and peroxidase activity. Furthermore, the dopamine groups on the shell surface facilitate folic acid modification, drug loading, and macrophage membrane camouflage. These functionalities collectively improve tumor-targeting abilities, promote immune evasion, and enhance tumor uptake. The doxorubicin-loaded MF-SPC (MDF-SPC) exhibited favorable dispersibility, stability, and pH-responsive sustained release properties. Both in vivo fluorescence imaging and NIR thermal imaging show that the MDF-SPC possessed active tumor-targeting capability. Thus, the MDF-SPC demonstrated high antitumor activity and biosafety when subjected to NIR laser irradiation in 4T1 tumor-bearing mice. Overall, the active tumor-targeting and retention capabilities, along with the rapid temperature elevations produced by PTT, allow the MDF-SPC to precisely and swiftly amplify chemodynamic therapy, gas therapy, and chemotherapy, providing a novel multimodal nanoplatform to promote antitumor therapy.
{"title":"A novel macrophage membrane-camouflaged ultra-small copper sulfide and photothermal-responsive nitric oxide donor nanocomposites for enhanced synergistic antitumor therapy","authors":"Haoyang Gong , Xiaonan Li , Xinru Feng , Dongjing Wang , Xinyi Zhang , Kai Wang , Yanchen Liu , Lu Xu , Xueyan Zhou , Yanzhuo Zhang","doi":"10.1016/j.colsurfb.2025.115373","DOIUrl":"10.1016/j.colsurfb.2025.115373","url":null,"abstract":"<div><div>Ultra-small copper sulfide (CuS) nanoparticles (NPs) possess exceptional photothermal performance, Fenton-like reaction activities, and multimodal imaging characteristics, while sodium nitroprusside serves as a clinical nitric oxide (NO) donor. A judicious combination of these two agents may facilitate the development of a novel multimodal synergistic antitumor strategy. In this context, we developed macrophage membrane-camouflaged and folic acid-conjugated nanocomposites (MF-SPC) that demonstrate dual responsiveness to near-infrared (NIR) laser and pH, aiming to enhance multimodal synergistic antitumor therapy. Within these biomimetic nanocomposites, sodium nitroprusside-doped Prussian blue NPs serve as the cores for inducing photothermal-responsive NO donation, while polydopamine layer embedded with CuS dots (sub-5 nm) act as the shells. This configuration not only enhances photothermal effects but also promotes Fenton-like reaction activity, glutathione depletion capabilities, and peroxidase activity. Furthermore, the dopamine groups on the shell surface facilitate folic acid modification, drug loading, and macrophage membrane camouflage. These functionalities collectively improve tumor-targeting abilities, promote immune evasion, and enhance tumor uptake. The doxorubicin-loaded MF-SPC (MDF-SPC) exhibited favorable dispersibility, stability, and pH-responsive sustained release properties. Both <em>in vivo</em> fluorescence imaging and NIR thermal imaging show that the MDF-SPC possessed active tumor-targeting capability. Thus, the MDF-SPC demonstrated high antitumor activity and biosafety when subjected to NIR laser irradiation in 4T1 tumor-bearing mice. Overall, the active tumor-targeting and retention capabilities, along with the rapid temperature elevations produced by PTT, allow the MDF-SPC to precisely and swiftly amplify chemodynamic therapy, gas therapy, and chemotherapy, providing a novel multimodal nanoplatform to promote antitumor therapy.</div></div>","PeriodicalId":279,"journal":{"name":"Colloids and Surfaces B: Biointerfaces","volume":"260 ","pages":"Article 115373"},"PeriodicalIF":5.6,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145799404","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}