首页 > 最新文献

Colloids and Surfaces B: Biointerfaces最新文献

英文 中文
Carbon quantum dot modified MoS2 mediating photodynamic and photothermal therapy in rapid inactivation of pathogenic microorganisms 碳量子点修饰mos2介导病原菌快速失活的光动力和光热治疗。
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-18 DOI: 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.
传统的抗菌策略面临着耐药性和环境污染等挑战。相比之下,基于纳米材料的光毒性疗法,包括光动力疗法(PDT)和光热疗法(PTT),在增强抗菌功效和减轻耐药性方面表现出希望。在这项工作中,研究了碳量子点修饰二硫化钼(CQDs@MoS2)复合材料在光动力和光热治疗中快速灭活病原微生物的潜力和有效性。在氙灯照射下,MoS2产生活性氧(ROS),破坏细菌膜和DNA。CQDs的加入在红外光下产生局部热,增强了复合材料的电荷分离,增加了ROS的产生,从而产生PTT效应,增强了光催化和抗菌活性。该方法在5 min内显著灭活革兰氏阳性金黄色葡萄球菌(S.aureus)和革兰氏阴性大肠杆菌(E. coli),并具有较高的稳定性。这些疗法的协同作用促进了有效的细菌灭活,为水消毒和生物医学应用提供了一个有前途的解决方案。
{"title":"Carbon quantum dot modified MoS2 mediating photodynamic and photothermal therapy in rapid inactivation of pathogenic microorganisms","authors":"Yuting Zhai ,&nbsp;Jiaxiu Liu ,&nbsp;Yangru Dai ,&nbsp;Tianyang Li ,&nbsp;Pingping Meng ,&nbsp;Dongjiang Yang ,&nbsp;Daohao Li ,&nbsp;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}
引用次数: 0
Applications of metal enhanced fluorescence for the detection and quantification of extracellular vesicles 金属增强荧光在细胞外囊泡检测和定量中的应用
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-18 DOI: 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.
疾病的早期检测可以显著改善患者的预后,但在初始阶段,许多生物标志物在体液中的浓度仍然极低,这对传统诊断技术构成了挑战。由于细胞外囊泡(ev)存在于血液、唾液和尿液等体液中,并且它们的分子载物反映了它们的细胞来源,因此它们已成为一种有前途的非侵入性生物标志物。然而,由于其低浓度和纳米级尺寸,对与疾病相关的电动汽车进行敏感和准确的检测具有挑战性。金属增强荧光(MEF)最近被应用于克服这些限制,通过金属纳米结构的局部表面等离子体共振放大标记ev的荧光信号。这种扩增使ev相关生物标志物的检测和定量具有更高的灵敏度,为早期疾病诊断提供了潜力。然而,迄今为止,很少有评论专门关注基于mef的电动汽车及其分子货物的检测和定量应用。本文综述了基于mef的EV分析平台的最新进展,包括纳米孔阵列、金属纳米岛和纳米阵列、三维基质中的金纳米粒子、金属有机框架和其他等离子体方法。虽然目前的综述已经讨论了MEF生物传感应用的使用,但本文特别关注基于MEF的EV检测,重点介绍了这些平台的设计原理、光学特性、优点和局限性。总的来说,MEF为增强EV检测提供了一个强有力的策略,弥合了实验分析和临床应用之间的差距。
{"title":"Applications of metal enhanced fluorescence for the detection and quantification of extracellular vesicles","authors":"Isabella Walker,&nbsp;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}
引用次数: 0
Multifunctional collagen hydrogel accelerates infected wound repair through photothermal disinfection and pro-angiogenic activity 多功能胶原水凝胶通过光热消毒和促血管生成活性加速感染伤口修复。
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-17 DOI: 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.
作为抵抗外界伤害的主要物理和免疫屏障,皮肤完整性经常受到创伤、疾病或医源性损伤的损害,导致病原微生物入侵和伤口感染。细菌抗生素耐药性和生物膜形成带来的不断升级的挑战突出了迫切需要将有效的抗菌功效与有效的组织再生调节协同结合的治疗系统。因此,本研究开发了一种集快速光热杀菌和促再生生物活性于一体的多功能水凝胶平台。我们设计了一种装载了聚集诱导发射(AIE)纳米粒子的I型重组人源胶原水凝胶(AIE@RHCI),实现了从液体到凝胶状态的快速紫外触发交联。该水凝胶具有适当的膨胀能力、可控的降解动力学和良好的机械性能,能够在运动过程中稳定地粘附动态变形的皮肤。在808 nm近红外(NIR)照射下,AIE组件的光热转换效率为39.7 %。体外研究证实,AIE@RHCI水凝胶在近红外照射下将耐甲氧西林金黄色葡萄球菌(MRSA)的生存力降低至22.2% %,同时实现79.8% %的生物膜抑制。同时,重组胶原基质促进血管内皮细胞迁移,加速血管形成,主要促血管生成基因VEGF和CD31显著上调。该集成平台将光热杀菌纳米颗粒与生物活性重组胶原支架协同结合,为抗生素耐药伤口感染的管理提供了一种新颖而有前途的治疗策略,在慢性和感染伤口护理中具有临床转化潜力。
{"title":"Multifunctional collagen hydrogel accelerates infected wound repair through photothermal disinfection and pro-angiogenic activity","authors":"Hao Shen ,&nbsp;Tong Li ,&nbsp;Xueying Chen ,&nbsp;Zhurun Fang ,&nbsp;Yan Xu ,&nbsp;Kai Zheng ,&nbsp;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}
引用次数: 0
pH-responsive phytochemical hydrogel loaded with antioxidant nanozymes for synergistic ROS scavenging and targeted inflammatory bowel disease therapy 负载抗氧化纳米酶的ph响应植物化学水凝胶,用于协同ROS清除和靶向炎症性肠病治疗。
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-17 DOI: 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.
炎症性肠病(IBD)由于其复杂的病理生理涉及氧化应激和慢性炎症,仍然是一个治疗挑战。在这里,我们开发了一种多功能水凝胶系统(CeO2@Rh-gel),该系统整合了大黄酸,一种天然抗炎剂,以及具有清除ROS纳米酶活性的CeO2 NPs,用于控释IBD治疗。CeO2@Rh-gel是通过自组装制造的,形成一个ph响应网络,保护胃中的有效载荷,同时使发炎的结肠能够控制释放。在体外,CeO2@Rh-gel表现出协同抗氧化作用,模仿SOD和CAT消除ROS,同时抑制lps刺激的巨噬细胞中的促炎细胞因子。在dss诱导的小鼠结肠炎模型中,口服CeO2@Rh-gel可显著减轻疾病严重程度,这可以通过体重减轻、结肠长度正常化和组织病理学改善来证明。在机械上,水凝胶恢复氧化还原稳态和减轻全身炎症。通过结合ROS清除,抗炎作用和可控递送,CeO2@Rh-gel为IBD的精确治疗提供了一个有希望的双重功能策略。
{"title":"pH-responsive phytochemical hydrogel loaded with antioxidant nanozymes for synergistic ROS scavenging and targeted inflammatory bowel disease therapy","authors":"Yunong Shi ,&nbsp;Miaosheng Tian ,&nbsp;Shuo Shi ,&nbsp;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}
引用次数: 0
Fabrication of immune-enhancing vesicles from reassembled yeast vacuolar membranes 重组酵母液泡膜制备免疫增强囊泡
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-17 DOI: 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.
酵母液泡最近成为一种很有前途的生物纳米药物递送材料,与传统的合成系统相比,它具有更好的稳定性和有效性。它们的膜在结构和组成上与哺乳动物细胞相似,具有良好的生物相容性和高效细胞相互作用的潜力。这些独特的性质使重组液泡(ReV)成为开发安全有效的治疗递送平台的有吸引力的候选者。本研究优化了液泡重组工艺,以提高给药性能。我们比较了两种方法:方法1,使用传统的长时间超声和过滤,方法2,采用优化的5分钟超声而不过滤。方法1产生的重组液泡(ReVMtd1)诱导了适度的TLR2表达,提示轻微的免疫启动,没有明显的炎症细胞因子激活。相反,方法2产生的重组液泡(ReVMtd2)表现出更好的免疫激活,显示出iNOS和TLR2的剂量依赖性上调。此外,ReVMtd2对柔红霉素(DNR)的包封效率为12.9 %,与天然液泡相当,并在12个月的时间内保持结构稳定性。这些发现突出了ReVMtd2作为一种强大的、生物相容性的、高效的药物传递系统的潜力,提供了增强的治疗性能和长期稳定性。
{"title":"Fabrication of immune-enhancing vesicles from reassembled yeast vacuolar membranes","authors":"Su-Min Lee ,&nbsp;Yunyoung Cho ,&nbsp;Jiwoo Lim ,&nbsp;Seungwoo Chung ,&nbsp;Ngoc-Tu Nguyen ,&nbsp;Yang-Hoon Kim ,&nbsp;Sang-Ho Park ,&nbsp;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}
引用次数: 0
A user-friendly multifunctional hydrogel spray with adjustable mechanical properties for hemostasis and infected wound healing 一种用户友好的多功能水凝胶喷雾剂,具有可调节的机械性能,用于止血和感染伤口愈合。
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-17 DOI: 10.1016/j.colsurfb.2025.115378
Mengyao Gao , Zongliang Wang , Zhaohui Tang , Peibiao Zhang
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.
外伤性大出血是目前国际急救医学领域的热点问题。传统的止血材料如纱布、海绵等存在着对不规则伤口黏附性差、黏附强度低等问题。现有的止血水凝胶也存在活性止血效率不足、界面粘连性和柔韧性难以平衡、缺乏功能协同等瓶颈,难以满足临床需求。为了解决这些挑战,本研究开发了一种自我修复的水凝胶喷雾,通过动态希夫碱反应将氧化右旋糖酐(ODex)与聚乙烯亚胺(PEI)混合,即可原位形成有效止血。得益于“醛-氨基”活性交联机制,该水凝胶喷雾具有快速凝胶化,在15 s内触发交联,粘附强度超过10 kPa,同时具有良好的自愈性能,适应动态和不规则伤口。聚乙烯亚胺的静电物理抗菌特性能有效抑制革兰氏阳性菌和革兰氏阴性菌,避免耐药风险。通过调节右旋糖酐的氧化程度,进一步优化了材料的止血-粘附协同作用,制备水凝胶工艺简单、性价比高、用户友好且可常温保存,具有良好的生物相容性。动物实验表明,该水凝胶喷雾剂能快速覆盖并粘附创面,有效止血,同时降低感染风险,促进创面愈合,为开发新一代临床可用的创伤止血材料提供创新策略和技术支持。
{"title":"A user-friendly multifunctional hydrogel spray with adjustable mechanical properties for hemostasis and infected wound healing","authors":"Mengyao Gao ,&nbsp;Zongliang Wang ,&nbsp;Zhaohui Tang ,&nbsp;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}
引用次数: 0
The cinnamaldehyde-thiosemicarbazone-zinc (II) complex induces apoptosis in CAL-27 cells 肉桂醛-硫代氨基脲-锌(II)复合物诱导CAL-27细胞凋亡
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-16 DOI: 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.
目的口腔鳞状细胞癌(OSCC)是一种影响口腔、颌部和面部的主要恶性肿瘤。在本研究中,我们合成了肉桂醛-硫代氨基脲-锌(II)配合物(CTZn)来抑制OSCC细胞的增殖。方法采用3-(4,5-二甲基噻唑-2-基)-2,5-二苯基溴化四唑(MTT)实验、细胞集落形成实验、伤口愈合/划痕实验和跨井侵袭实验研究CTZn对OSCC的抑制作用。流式细胞术(FCM)探讨CTZn对细胞凋亡和细胞周期进程的影响。通过体内实验,评价其在OSCC异种移植小鼠模型和原位肿瘤模型小鼠中的抗肿瘤活性。通过检测靶向代谢物,进行药物亲和力响应性靶稳定性(dart)实验、分子对接分析和Western blot分析,探索CTZn的作用机制。还检测了CAL-27细胞中活性氧(ROS)、氧化和还原的烟酰胺腺嘌呤二核苷酸磷酸(NADP + /NADPH)、谷胱甘肽(GSH)和6-磷酸葡萄糖酸脱氢酶(PGD)蛋白的水平。结果CTZn体外抑制CAL-27细胞生长呈时间和浓度依赖性,IC50值为1.642 ~ 2.223μmol/L,体内也具有抗肿瘤活性。CTZn还能抑制PGD信使RNA (mRNA)和蛋白表达,降低NADP+ /NADPH和GSH水平,显著增加ROS水平,从而诱导氧化应激。结论ctzn可损伤CAL-27细胞线粒体功能,降低ATP水平,诱导G₂相阻滞和细胞凋亡。因此,它是治疗OSCC的理想药物。
{"title":"The cinnamaldehyde-thiosemicarbazone-zinc (II) complex induces apoptosis in CAL-27 cells","authors":"Chen-yan Li ,&nbsp;Shuohua Xie ,&nbsp;Min Deng ,&nbsp;Ganrong Huang ,&nbsp;Yanqiang Huang ,&nbsp;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}
引用次数: 0
The MCM/Lys-Cys nanodevices for the efficient gene delivery: An approach towards MCP1 gene manipulation using CRISPR technology 用于高效基因传递的MCM/Lys-Cys纳米器件:使用CRISPR技术操作smcp1基因的方法。
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-16 DOI: 10.1016/j.colsurfb.2025.115377
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
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.
乳腺癌仍然是全世界妇女中最常见的恶性肿瘤,需要新的治疗方法。本研究研究了一种创新的基因递送策略,利用赖氨酸和半胱氨酸修饰的介孔二氧化硅纳米颗粒(MCM-41)有效递送靶向单核细胞趋化蛋白-1 (MCP-1/CCL2)基因的CRISPR-Cas9质粒。TCGA-BRCA数据集的生物信息学分析揭示了CCL2在乳腺癌中的显著失调,强调其参与肿瘤生长和炎症。MCM/Lys-Cys纳米载体表现出卓越的生物相容性,并有效地封装了含有GFP的质粒,与传统技术相比,促进了MDA-MB-231乳腺癌细胞的细胞摄取。功能实验表明,CRISPR/ cas9介导的CCL2抑制可显著降低细胞增殖、迁移和侵袭,突出了这种靶向基因治疗策略在乳腺癌治疗中的前景。这些发现表明,MCM/Lys-Cys纳米系统为精确的基因编辑提供了一种可行的非病毒方法,可能通过调节炎症途径促进对乳腺癌的治疗。
{"title":"The MCM/Lys-Cys nanodevices for the efficient gene delivery: An approach towards MCP1 gene manipulation using CRISPR technology","authors":"Azadeh Rahimi ,&nbsp;Ilnaz Rahimmanesh ,&nbsp;Navid Abedpoor ,&nbsp;Maryam Boshtam ,&nbsp;Elham Bidram ,&nbsp;Shaghayegh Haghjooy Javanmard ,&nbsp;Hossein Khanahmad ,&nbsp;Laleh Rafiee ,&nbsp;Ashkan Bigham ,&nbsp;Mohammad Rafienia ,&nbsp;Saeed Karbasi ,&nbsp;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}
引用次数: 0
Fighting periodontitis with a pH-triggered nanocoating: A sustained-release strategy for simvastatin delivery 用ph触发的纳米涂层对抗牙周炎:辛伐他汀递送的缓释策略。
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-15 DOI: 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.
牙周炎的发病率不断增加,需要创新的治疗方法,因为目前的药物治疗受到保留时间短、清除速度快和抗菌素耐药性风险的限制。本研究介绍了一种新型的辛伐他汀(SIM) ph响应纳米胶系统。其核心创新在于成为同类系统中第一个通过交联“外壳”稳定的系统((SIM NP)n),这大大提高了其稳定性和控制释放能力。这种“智能”系统在中性环境(pH 7.4)中保持稳定,防止药物过早释放。然而,当遇到牙周炎的酸性炎症微环境时,外壳降解,直接在炎症部位触发SIM的持续释放。体外研究表明,载sim胶束具有强大的免疫调节作用:能有效抑制促炎M1巨噬细胞极化(降低IL-1β、iNOS),促进抗炎M2巨噬细胞极化(增加Arg-1、IL-10)。机制研究证实这种治疗作用是由PI3K/AKT/mTOR信号通路介导的。更重要的是,在牙周炎小鼠模型中,纳米胶束显著减少了牙槽骨吸收,在体内显示出强大的抗炎和骨保护功效。总之,这种“智能”ph触发的SIM递送系统为克服传统治疗方法的局限性提供了一种非常有前途和有效的策略,为牙周炎的治疗提供了新的靶向途径。
{"title":"Fighting periodontitis with a pH-triggered nanocoating: A sustained-release strategy for simvastatin delivery","authors":"Siyu Du ,&nbsp;Yi Zheng ,&nbsp;Yabing Chen ,&nbsp;Yijing Guo ,&nbsp;Xinyue Kang ,&nbsp;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}
引用次数: 0
A novel macrophage membrane-camouflaged ultra-small copper sulfide and photothermal-responsive nitric oxide donor nanocomposites for enhanced synergistic antitumor therapy 一种新型巨噬细胞膜伪装的超小硫化铜和光热响应型一氧化氮供体纳米复合材料,用于增强协同抗肿瘤治疗
IF 5.6 2区 医学 Q1 BIOPHYSICS Pub Date : 2025-12-15 DOI: 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.
超小硫化铜纳米颗粒(cu)具有优异的光热性能、芬顿样反应活性和多模态成像特性,而硝普钠可作为临床一氧化氮(NO)供体。这两种药物的明智组合可能促进一种新的多模式协同抗肿瘤策略的发展。在此背景下,我们开发了巨噬细胞膜伪装和叶酸共轭纳米复合材料(MF-SPC),其对近红外(NIR)激光和pH值具有双重响应性,旨在增强多模态协同抗肿瘤治疗。在这些仿生纳米复合材料中,硝普钠掺杂的普鲁士蓝NPs作为诱导光热响应性NO捐赠的核心,而嵌入cu点(亚5 nm)的聚多巴胺层作为壳层。这种结构不仅增强了光热效应,而且促进了芬顿样反应活性、谷胱甘肽耗竭能力和过氧化物酶活性。此外,壳表面的多巴胺基团促进叶酸修饰,药物装载和巨噬细胞膜伪装。这些功能共同提高肿瘤靶向能力,促进免疫逃避,增强肿瘤摄取。负载多柔比星的MF-SPC (MDF-SPC)具有良好的分散性、稳定性和ph响应缓释性能。体内荧光成像和近红外热成像均显示MDF-SPC具有活跃的肿瘤靶向能力。因此,MDF-SPC在4T1荷瘤小鼠近红外激光照射下表现出较高的抗肿瘤活性和生物安全性。总的来说,活跃的肿瘤靶向和保留能力,以及PTT产生的快速温度升高,使MDF-SPC能够精确、快速地放大化学动力学治疗、气体治疗和化疗,为促进抗肿瘤治疗提供了一种新的多模态纳米平台。
{"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 ,&nbsp;Xiaonan Li ,&nbsp;Xinru Feng ,&nbsp;Dongjing Wang ,&nbsp;Xinyi Zhang ,&nbsp;Kai Wang ,&nbsp;Yanchen Liu ,&nbsp;Lu Xu ,&nbsp;Xueyan Zhou ,&nbsp;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}
引用次数: 0
期刊
Colloids and Surfaces B: Biointerfaces
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1