ACS Applied Bio Materials最新文献

筛选
英文 中文
Apoptosis-Inducing Effect of Cytocompatible Poly(Acrylic Acid)-Coated Temperature-Sensitive Mn0.9Zn0.1Fe2O4 Magnetic Fluid on Breast Cancer Cells MCF-7 细胞相容性聚丙烯酸包被温敏Mn0.9Zn0.1Fe2O4磁流体对乳腺癌细胞MCF-7的诱导凋亡作用
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-25 DOI: 10.1021/acsabm.5c01075
Tushar Patel, Kinnari Parekh* and Neeraj Jain*, 
{"title":"Apoptosis-Inducing Effect of Cytocompatible Poly(Acrylic Acid)-Coated Temperature-Sensitive Mn0.9Zn0.1Fe2O4 Magnetic Fluid on Breast Cancer Cells MCF-7","authors":"Tushar Patel,&nbsp;Kinnari Parekh* and Neeraj Jain*,&nbsp;","doi":"10.1021/acsabm.5c01075","DOIUrl":"10.1021/acsabm.5c01075","url":null,"abstract":"<p >Cancer remains a challenging disease to treat due to the limitations of conventional therapies, including toxicity to healthy cells and drug resistance. This underscores the need for alternative treatment modalities to target tumors more effectively while minimizing side effects. Magnetic nanoparticles (MNPs) have emerged as promising agents in the realm of cancer therapy, particularly through magnetic fluid hyperthermia (MFH), which employs MNPs to generate localized heat in tumor tissues using an alternating magnetic field (AMF), while sparing healthy cells. This present study focused on the synthesis of self-regulating, temperature-controlled, poly(acrylic acid) (PAA)-coated manganese–zinc ferrite nanoparticle-based magnetic fluid (MF) to enhance the biocompatibility of MF and its therapeutic efficacy. The biocompatibility and cellular uptake of the synthesized MF were investigated by using the MTT assay and Prussian blue staining. Quantitative assessment of the internalization of MNPs was carried out using inductively coupled plasma-mass spectrometry (ICP-MS). The study further analyzed the immediate and 24 h delayed cytotoxic effects of MFH on breast cancer cells (MCF-7) and noncancerous murine fibroblasts (McCoy). The mode of cell death induced immediately and after 24 h post-MFH was examined using Hoechst/propidium iodide staining and flow cytometry. Both MCF-7 and McCoy cells exhibited favorable biocompatibility and substantial internalization of MF, with a significant induction of apoptosis after MFH. A 24 h incubation period post-MFH enabled cells to transition from early- to late-stage apoptosis in both cell lines, resulting in nearly complete cell death. This research establishes a foundational framework for investigating the application of PAA-coated biocompatible MNPs, with the goal of further exploring their use in breast cancer spheroids and/or in vivo animal models to develop MFH-based cancer treatment strategies.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":"8 9","pages":"8101–8114"},"PeriodicalIF":4.7,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935754","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chondroitin Sulfate-Mediated Targeted Delivery of Docetaxel and Silibinin-Loaded Nanoparticles for Amelioration of Anticancer Efficacy in Breast Cancer 硫酸软骨素介导的靶向递送多西紫杉醇和水飞蓟宾纳米颗粒改善乳腺癌的抗癌效果。
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-25 DOI: 10.1021/acsabm.5c01098
Mayur Aalhate, Srushti Mahajan, Rahul Kumar, Arun Kumar, Barkha Jangir, Sharon Munagalasetty, Umarani Brahma, Ujala Gupta, Indrani Maji, Durga P. Mishra, Vasundhra Bhandari and Pankaj Kumar Singh*, 
{"title":"Chondroitin Sulfate-Mediated Targeted Delivery of Docetaxel and Silibinin-Loaded Nanoparticles for Amelioration of Anticancer Efficacy in Breast Cancer","authors":"Mayur Aalhate,&nbsp;Srushti Mahajan,&nbsp;Rahul Kumar,&nbsp;Arun Kumar,&nbsp;Barkha Jangir,&nbsp;Sharon Munagalasetty,&nbsp;Umarani Brahma,&nbsp;Ujala Gupta,&nbsp;Indrani Maji,&nbsp;Durga P. Mishra,&nbsp;Vasundhra Bhandari and Pankaj Kumar Singh*,&nbsp;","doi":"10.1021/acsabm.5c01098","DOIUrl":"10.1021/acsabm.5c01098","url":null,"abstract":"<p >Breast cancer is the second leading cause of mortality in women worldwide. Chemotherapeutic drugs like docetaxel (DTX) remain key molecules in cancer management. Silibinin (SLB) is an effective agent causing apoptosis and autophagy resulting in cancer cell death. Recently, ligand-anchored targeted nanocarrier-based drug delivery has achieved substantial improvement in cancer therapy. In the present study, chondroitin sulfate (CS) was used as a ligand to target CD44 receptors overexpressed in breast cancer cells. Herein, CS-coated chitosan–lecithin nanoparticles (CS-DTX-SLB-LCNPs) were developed for co-delivery of DTX and SLB. The CS-DTX-SLB-LCNP resulted in a particle size of 208.33 ± 2.20 nm with an entrapment efficiency of 83.81% for DTX and 92.96% for SLB. Further, the dialysis release study showed sustained release behavior, and a hemocompatible nature which was proved by the hemolysis study. The cell cytotoxicity in MDA-MB-231 cells revealed considerably higher cell killing with CS-DTX-SLB-LCNP compared to free drugs. The cell uptake studies showed a 1.97-fold and 2.45-fold rise in fluorescence intensity from C6-LCNP and CS-C6-LCNP, respectively, as compared to free C6. CS-DTX-SLB-LCNP caused a sharp rise in the ROS level and resulted in mitochondrial membrane depolarization, which induced apoptosis and cell death. An <i>in vivo</i> efficacy study in Balb/c mice demonstrated 2.24-fold and 2-fold reduction in tumor volume after CS-DTX-SLB-LCNP treatment as compared to free DTX and free SLB groups. In conclusion, CS-DTX-SLB-LCNPs showed encouraging prospects in increasing cellular uptake and targeting specificity to treat breast cancer.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":"8 9","pages":"8126–8145"},"PeriodicalIF":4.7,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935757","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Photo-Activated Cu-ZIF-8 Integrated Sprayable Hydrogels for Accelerated Wound Healing 光活化Cu-ZIF-8集成可喷雾水凝胶加速伤口愈合。
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-25 DOI: 10.1021/acsabm.5c01127
Li Li, Yueming Zhu, Xurui Li, Dongyu Li, Kai Jia and Yu Liu*, 
{"title":"Photo-Activated Cu-ZIF-8 Integrated Sprayable Hydrogels for Accelerated Wound Healing","authors":"Li Li,&nbsp;Yueming Zhu,&nbsp;Xurui Li,&nbsp;Dongyu Li,&nbsp;Kai Jia and Yu Liu*,&nbsp;","doi":"10.1021/acsabm.5c01127","DOIUrl":"10.1021/acsabm.5c01127","url":null,"abstract":"<p >To address the growing challenge of antibiotic-resistant infections in wound healing, we developed a multifunctional hydrogel dressing (Nar@Cu<sub>15%</sub>ZIF-8@Hydrogel) combining the natural flavonoid naringenin (Nar) with copper-doped ZIF-8 nanoparticles. Nar provides anti-inflammatory and proangiogenic effects, while Cu-doped ZIF-8 enhances reactive oxygen species (ROS) production under near-infrared (NIR) light, enabling synergistic photodynamic and photothermal antibacterial therapy. These particles were embedded in a thermosensitive poloxamer hydrogel, offering excellent biocompatibility, sprayability, and in situ gelation. The composite dressing achieved over 90% antibacterial efficiency against <i>Escherichia coli</i> and <i>Staphylococcus aureus</i>, significantly promoted cell migration (66.7% at 24 h), and showed low cytotoxicity. In vivo, it accelerated wound healing (99.91% on day 10), reduced inflammation (increased tumor necrosis factor-α (TNF-α) expression), and enhanced angiogenesis (CD31 fluorescence up to 154.7). This rationally designed dressing combines targeted antibacterial action with regenerative capacity, presenting a promising strategy for treating infected wounds.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":"8 9","pages":"8197–8217"},"PeriodicalIF":4.7,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeted Delivery of Bezafibrate via Silica Nanoparticles Restores Mitochondrial Function and Reduces Oxidative Stress in Insulin-Resistant Cells. 通过二氧化硅纳米颗粒靶向递送bezafbrate可恢复线粒体功能并减少胰岛素抵抗细胞的氧化应激。
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-25 DOI: 10.1021/acsabm.5c00721
Arooma Jannat, Shanmugavel Chinnathambi, Gopinath Packirisamy, Leon Bernet Shano, Karthikeyan Subramani, Rajendiran Mangaiyarkarasi, Yuichi Taniguchi, Ganesh N Pandian
{"title":"Targeted Delivery of Bezafibrate via Silica Nanoparticles Restores Mitochondrial Function and Reduces Oxidative Stress in Insulin-Resistant Cells.","authors":"Arooma Jannat, Shanmugavel Chinnathambi, Gopinath Packirisamy, Leon Bernet Shano, Karthikeyan Subramani, Rajendiran Mangaiyarkarasi, Yuichi Taniguchi, Ganesh N Pandian","doi":"10.1021/acsabm.5c00721","DOIUrl":"https://doi.org/10.1021/acsabm.5c00721","url":null,"abstract":"<p><p>Mitochondrial dysfunction and oxidative stress are pivotal drivers of obesity-induced insulin resistance, posing significant challenges to therapeutic efficacy. Bezafibrate, a pan-peroxisome proliferator-activated receptor (PPAR) agonist, enhances mitochondrial metabolism and antioxidant defenses; however, its efficacy is hindered by poor solubility and bioavailability. In this study, we engineered biodegradable periodic mesoporous organosilica (BPMO) nanoparticles to improve bezafibrate delivery and intracellular efficacy. Spectroscopic, circular dichroism, and molecular modeling analyses confirmed that bezafibrate stably binds antioxidant enzymes catalase and superoxide dismutase (SOD), with minimal perturbation to their conformation. Molecular docking and dynamics simulations supported these findings by demonstrating stable binding and increased protein structural integrity. In insulin-resistant human adipose-derived cells (HPAd), BPMO-bezafibrate notably restored mitochondrial membrane potential, enhanced fatty acid oxidation, reduced intracellular reactive oxygen species (ROS), and upregulated endogenous gene expression of PPARγ and adiponectin. Compared to liposomal and free-drug delivery, BPMO-bezafibrate showed higher cellular uptake, reduced cytotoxicity, and improved metabolic rescue. Moreover, in vivo uptake and safety were validated using medaka embryos. Collectively, our findings establish BPMO-assisted bezafibrate delivery as a safe and potent strategy to restore mitochondrial function and redox balance in insulin-resistant cells, offering translational promise for treating metabolic disorders.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935606","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development of a Hydrogel Platform with GBM and Microglia: A Potential Glioblastoma Tumor Model 胶质母细胞瘤和小胶质细胞的水凝胶平台的开发:一种潜在的胶质母细胞瘤模型。
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-25 DOI: 10.1021/acsabm.5c00735
Seyma Isik, Deniz Yucel and Vasif Hasirci*, 
{"title":"Development of a Hydrogel Platform with GBM and Microglia: A Potential Glioblastoma Tumor Model","authors":"Seyma Isik,&nbsp;Deniz Yucel and Vasif Hasirci*,&nbsp;","doi":"10.1021/acsabm.5c00735","DOIUrl":"10.1021/acsabm.5c00735","url":null,"abstract":"<p >Glioblastoma (GBM) is an aggressive brain tumor with a complex microenvironment shaped by a dense extracellular matrix (ECM) and dynamic interactions with stromal cells, presenting major challenges for <i>in vitro</i> modeling. In this study, we developed a biomimetic hydrogel platform by integrating a brain-derived decellularized extracellular matrix (dECM) with hyaluronic acid methacrylate (HAMA), yielding a composite (1H3D) that closely reflects the ECM characteristics of GBM tissue. Mechanically, 1H3D hydrogels exhibited a compressive modulus of 9.44 ± 0.73 kPa and an elastic modulus of 458.30 ± 13.39 Pa, resembling native GBM tissue. By retaining biochemical components from the brain dECM, hydrogels support key cellular processes such as adhesion, matrix remodeling, and invasion. These functions are essential for mimicking the highly invasive, plastic, and adaptive behavior of glioblastoma, thereby enhancing the physiological relevance of the <i>in vitro</i> platform. Coculture with microglia promoted glioblastoma progression, as evidenced by a 43% increase in <i>K</i><sub>i</sub>-67 expression and a 41% increase in invasion distance, underscoring the protumoral role of microglia–glioblastoma interactions within the engineered microenvironment. Altogether, integration of a GBM relevant hydrogel matrix with microglia coculture provides a biologically and mechanically representative <i>in vitro</i> platform that reproduces key features of tumor–stroma interactions, offering a useful tool for studying glioblastoma progression and enhancing the translational potential of preclinical models.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":"8 9","pages":"7757–7770"},"PeriodicalIF":4.7,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsabm.5c00735","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935712","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Graphene Oxide Facilitates Osteo-Differentiation of Mesenchymal Stem Cells in 3-Dimensional Porous Vitreous Carbon Foam Scaffolds 氧化石墨烯促进三维多孔玻璃碳泡沫支架中间充质干细胞的骨分化。
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-24 DOI: 10.1021/acsabm.5c00996
Rohit Bagal, Manjushree Bahir, Nibedita Lenka* and T. Umasankar Patro*, 
{"title":"Graphene Oxide Facilitates Osteo-Differentiation of Mesenchymal Stem Cells in 3-Dimensional Porous Vitreous Carbon Foam Scaffolds","authors":"Rohit Bagal,&nbsp;Manjushree Bahir,&nbsp;Nibedita Lenka* and T. Umasankar Patro*,&nbsp;","doi":"10.1021/acsabm.5c00996","DOIUrl":"10.1021/acsabm.5c00996","url":null,"abstract":"<p >Incorporation of nanofillers into carbon foam (CF) is an amenable process to enhance its biological properties, which otherwise is bioinert. In the present study, CF-reinforced graphene oxide (CFGO) was considered to study the effects of GO on the structure, <i>in vitro</i> bioactivity, and biocompatibility using gingival mesenchymal stem cells (gMSCs) as the cellular source. CF was prepared by carbonization of polyurethane (PU) foam, and for the synthesis of GO-incorporated PU foam, GO was dispersed in isocyanate, one of the constituents of PU foam. The bioactivity of the CFGO composite scaffolds with GO content ranging from 0.31 to 0.94 wt % to PU foam was examined using simulated body fluid (SBF). Notwithstanding the GO addition, the CF scaffolds showed apatite growth on the scaffold surface, and the apatite layer increased with an increase in GO content in CF, as ascertained by X-ray diffraction (XRD), scanning electron microscopy (SEM), Fourier-transform infrared spectroscopy (FTIR), and elemental analysis using energy dispersive X-ray spectroscopy (EDS). The <i>in vitro</i> biocompatibility of the CF scaffolds, examined using the methyl thiazole tetrazolium (MTT) assay, revealed a proportionate increment in gMSC viability with increased GO content in the scaffolds. Additionally, GO promoted the attachment, growth, and infiltration of gMSCs across the CFGO scaffolds, suggesting its involvement in improving bioactivity and biocompatibility properties. Interestingly, the osteocyte differentiation of gMSCs was also enhanced with an increased GO concentration in the scaffolds. Together, our findings suggest that the CFGO composite scaffold can serve as a promising candidate for bone tissue engineering.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":"8 9","pages":"8010–8021"},"PeriodicalIF":4.7,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935750","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Heavy-Atom-Free Aza-BODIPY Polymeric Nanoparticles for Theranostic-Guided Photodynamic Cancer Therapy 用于治疗导向光动力癌症治疗的无重原子Aza-BODIPY聚合纳米颗粒。
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-21 DOI: 10.1021/acsabm.5c00878
Prapassara Muangsopa, Gong Yi Yong, Yuranan Thathong, Sastiya Kampaengsri, Kantapat Chansaenpak, Jadsada Saetiew, Tunyawat Khrootkaew, Ailada Jantasin, Panomsak Meemon, Mongkol Sukwattanasinitt, Chin Siang Kue* and Anyanee Kamkaew*, 
{"title":"Heavy-Atom-Free Aza-BODIPY Polymeric Nanoparticles for Theranostic-Guided Photodynamic Cancer Therapy","authors":"Prapassara Muangsopa,&nbsp;Gong Yi Yong,&nbsp;Yuranan Thathong,&nbsp;Sastiya Kampaengsri,&nbsp;Kantapat Chansaenpak,&nbsp;Jadsada Saetiew,&nbsp;Tunyawat Khrootkaew,&nbsp;Ailada Jantasin,&nbsp;Panomsak Meemon,&nbsp;Mongkol Sukwattanasinitt,&nbsp;Chin Siang Kue* and Anyanee Kamkaew*,&nbsp;","doi":"10.1021/acsabm.5c00878","DOIUrl":"10.1021/acsabm.5c00878","url":null,"abstract":"<p >Photodynamic therapy (PDT) presents a noninvasive method for cancer treatment, characterized by high spatial selectivity and reduced side effects. However, conventional photosensitizers containing heavy atoms often pose risks of cellular toxicity, limiting their clinical translation. To address this, we developed a heavy-atom-free aza-BODIPY derivative (<b>AB–OH</b>) with a donor–acceptor–donor (D–A–D) structure, conjugated to poly(ethylene glycol)-succinic acid (mPEG-SA) to form amphiphilic <b>mPEG-SA-AB</b>. This conjugate self-assembles into stable nanoparticles (NPs) that can generate reactive oxygen species (ROS) and produce mild heat upon irradiation with 660 nm red light from an LED. <i>In vitro</i> studies on 4T1 breast cancer cells revealed that <b>mPEG-SA-AB</b> NPs maintain high biocompatibility under dark conditions and induce significant photocytotoxicity upon irradiation, as confirmed by intracellular ROS detection, live/dead staining assays, and apoptosis by flow cytometry. Confocal microscopy revealed increased cellular uptake and accumulation of NPs in endocytic compartments in both 2D and 3D culture models, compared to the free dye. Moreover, optical coherence tomography (OCT) enabled real-time, noninvasive monitoring of PDT efficacy in 3D cultures, supporting the platform’s translational potential. Finally, <i>in vivo</i> fluorescence imaging in 4T1 tumor-bearing BALB/c mice confirmed the targeted tumor accumulation and superior antitumor performance of the NPs, achieving 2.5-fold greater therapeutic efficacy than the parent <b>AB–OH</b> molecule. The NPs also exhibited excellent hemocompatibility and systemic biocompatibility. Overall, <b>mPEG-SA-AB</b> NPs represent a safe and potent nanotheranostic platform, offering synergistic photodynamic and mild photothermal effects for enhanced cancer diagnosis and therapy.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":"8 9","pages":"7842–7854"},"PeriodicalIF":4.7,"publicationDate":"2025-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsabm.5c00878","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935747","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Poly(VP-co-V3D3), an Insoluble Hydrophilic Copolymer Coating Deposited via Initiated Chemical Vapor Deposition to Prevent Bacterial Adhesion and Biofilm Formation 聚(VP-co-V3D3),一种通过化学气相沉积沉积的不溶性亲水性共聚物涂层,可防止细菌粘附和生物膜的形成。
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-21 DOI: 10.1021/acsabm.5c00974
Amelia Whiteley, Guillaume Nonglaton* and Vincent Jousseaume, 
{"title":"Poly(VP-co-V3D3), an Insoluble Hydrophilic Copolymer Coating Deposited via Initiated Chemical Vapor Deposition to Prevent Bacterial Adhesion and Biofilm Formation","authors":"Amelia Whiteley,&nbsp;Guillaume Nonglaton* and Vincent Jousseaume,&nbsp;","doi":"10.1021/acsabm.5c00974","DOIUrl":"10.1021/acsabm.5c00974","url":null,"abstract":"<p >While bacterial biofilm remains one of the world’s major health issues, many traditional strategies for combatting it are being questioned. Indeed, as antibioresistance is rising and other biocides are raising concerns due to their potential toxicity, these curative methods no longer seem to be the solution. Therefore, biomimetic preventive methods, based on impeding bacterial adhesion, are being developed. One strategy is creating superhydrophilic surfaces to which bacteria cannot adhere as a water barrier forms at the interface. Polymer thin films are an efficient way for crafting such surfaces, and more precisely, the vacuum-based method initiated chemical vapor deposition (iCVD) allows the formation of polymers through radical polymerization mechanisms, therefore giving good control over the chemical composition of the resulting coatings. Polyvinylpyrrolidone (poly(VP)) has previously been used for its antibiofouling properties due to its high wettability and can be easily deposited into thin films by iCVD. However, its strong affinity with water makes it soluble, and thus, it cannot be used in bacterial suspensions. By coinjecting the VP precursor with a cross-linker, in this case organosilicate 1,3,5-trivinyl-1,3,5-trimethylcyclotrisiloxane (V3D3), the iCVD technique allows the formation of cross-linked copolymer thin films. The conservation of the functional groups of both monomers, whose proportion can be finely tuned by adjusting the injected fractions of each precursor, was studied by Fourier transform infrared spectroscopy (FTIR) and X-ray photoelectron spectroscopy (XPS). The Fineman–Ross equation was used to evaluate the reactivity of both monomers, allowing us to describe the copolymer structure. Water contact angle (WCA) and atomic force microscopy (AFM) measurements were carried out to characterize the surface state of the coatings and correlate it with the copolymer composition. Finally, solubility tests were carried out in phosphate buffer saline (PBS), and microbiological adhesion assays were carried out with the model species <i>Escherichia coli</i> to highlight the potential of these copolymers for antifouling applications.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":"8 9","pages":"7979–7988"},"PeriodicalIF":4.7,"publicationDate":"2025-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935616","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Conjugation of TLR7/8 Adjuvants with Cholesteryl Pullulan for a Self-Assembled Nanogel Vaccine: Enhanced Immunostimulatory Activation with Reduced Systemic Inflammation TLR7/8佐剂与胆固醇普鲁兰偶联制备自组装纳米凝胶疫苗:增强免疫刺激激活和减少全身炎症
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-21 DOI: 10.1021/acsabm.5c00866
Yuki Yazawa, Shin-ichi Sawada, Yoshihiro Sasaki* and Kazunari Akiyoshi*, 
{"title":"Conjugation of TLR7/8 Adjuvants with Cholesteryl Pullulan for a Self-Assembled Nanogel Vaccine: Enhanced Immunostimulatory Activation with Reduced Systemic Inflammation","authors":"Yuki Yazawa,&nbsp;Shin-ichi Sawada,&nbsp;Yoshihiro Sasaki* and Kazunari Akiyoshi*,&nbsp;","doi":"10.1021/acsabm.5c00866","DOIUrl":"10.1021/acsabm.5c00866","url":null,"abstract":"<p >Therapeutic cancer vaccines are promising treatment strategies capable of specifically eradicating cancer cells. However, their clinical efficacy has been limited due to insufficient cytotoxic T lymphocyte (CTL) responses. Effective CTL induction requires efficient and proper coordination of immune interactions between antigen-presenting cells (APCs) and T cells within lymphoid tissues. Here, we developed a cholesteryl pullulan (CHP) nanogel carrier system that codelivers protein antigens and Toll-like receptor 7/8 (TLR7/8) adjuvants. In this system, the TLR7/8 adjuvants are covalently conjugated to cholesteryl pullulan, while the protein antigens are noncovalently incorporated with the nanogel through host–guest interactions. This dual incorporation strategy enables the simultaneous delivery of both components to lymphoid tissues, enhancing CTL responses while minimizing systemic toxicity. Owing to its small size (approximately 25 nm) and neutral surface charge, the nanogel vaccine efficiently migrated to the lymph nodes after subcutaneous administration, delivering both the antigens and adjuvants to APCs. The nanogel system effectively activated APCs in the draining lymph nodes while minimizing systemic inflammatory responses by restricting adjuvant diffusion at the injection site. This system led to the robust induction of antigen-specific CTLs. Overall, we demonstrated that the adjuvant-conjugated nanogel vaccine effectively controlled the pharmacokinetics of both antigens and adjuvants, resulting in enhanced CTL induction. These findings provide valuable insights for designing safer and more effective vaccines, contributing to advancements in cancer immunotherapy.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":"8 9","pages":"7899–7908"},"PeriodicalIF":4.7,"publicationDate":"2025-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935682","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Injectable Antimicrobial Quaternary Chitosan Salt/Tannic Acid as a Delivery Platform for Enhanced Cranial Bone Regeneration via the Synergy of BMP-2 and VEGF 通过BMP-2和VEGF协同作用增强颅骨再生的注射用抗菌季铵盐/单宁酸递送平台
IF 4.7
ACS Applied Bio Materials Pub Date : 2025-08-21 DOI: 10.1021/acsabm.5c00935
Zeyu Liu, Yanxin Liu, Tongyu Shi* and Kun Fu*, 
{"title":"Injectable Antimicrobial Quaternary Chitosan Salt/Tannic Acid as a Delivery Platform for Enhanced Cranial Bone Regeneration via the Synergy of BMP-2 and VEGF","authors":"Zeyu Liu,&nbsp;Yanxin Liu,&nbsp;Tongyu Shi* and Kun Fu*,&nbsp;","doi":"10.1021/acsabm.5c00935","DOIUrl":"10.1021/acsabm.5c00935","url":null,"abstract":"<p >Treating large, irregularly shaped bone defects continues to be a major challenge in clinical practice. In this work, an innovative injectable antimicrobial delivery platform is introduced. The therapeutic system leverages the combination of quaternary chitosan salt (QCS) and tannic acid (TA), synergistically supplemented with BMP-2 and VEGF, to enhance the regenerative efficacy. Hydrogen bonding between QCS and TA enhances mechanical strength, while the localized delivery of BMP-2 and VEGF promotes osteogenesis and angiogenesis. The sustained antimicrobial activity of QCS/TA further supports a regenerative microenvironment. Mechanistic analysis showed upregulated osteogenic gene expression in the QCS/TA/BMP-2/VEGF group, consistent with <i>in vivo</i> results─nearly 80% defect closure within 8 weeks. The injectable QCS/TA hydrogel platform incorporated with BMP-2 and VEGF represents a promising strategy for repairing bone defects, emphasizing its pioneering role in addressing clinical challenges.</p>","PeriodicalId":2,"journal":{"name":"ACS Applied Bio Materials","volume":"8 9","pages":"7909–7924"},"PeriodicalIF":4.7,"publicationDate":"2025-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935735","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信