Seong-Jin Kim, In Ho Kang, Hyuk Joo Ahn, Wan Jin Cho, Hyun Jung Kim, Jinho Shin, Min Kyu Sung, Jae Hoon Lee, Eunsung Jun
{"title":"Hemostatic and adhesion prevention performance of an extracellular matrix based novel agent in a mouse liver laceration model.","authors":"Seong-Jin Kim, In Ho Kang, Hyuk Joo Ahn, Wan Jin Cho, Hyun Jung Kim, Jinho Shin, Min Kyu Sung, Jae Hoon Lee, Eunsung Jun","doi":"10.1088/1748-605X/ae0d21","DOIUrl":"https://doi.org/10.1088/1748-605X/ae0d21","url":null,"abstract":"<p><p>Traumatic bleeding and tissue damage pose complex clinical challenges requiring rapid hemostasis and concurrent tissue regeneration. Although traditional hemostatic agents primarily focus on controlling bleeding, they generally lack additional functionalities such as preventing adhesion and promoting tissue regeneration, limiting their clinical utility. This study developed a composite regenerative hemostatic agent based on a porcine decellularized extracellular matrix (ECM) to address these limitations. This agent is designed to achieve rapid hemostasis, prevent adhesions, and promote tissue regeneration. Its functionality was evaluated using a mouse liver laceration model to explore its clinical applicability. Hemostatic efficacy was assessed by measuring the bleeding time and blood loss, and comparing the composite agent with conventional commercial hemostatic agents. Additionally, the degree of adhesion between the liver and surrounding tissues was evaluated after re-opening the abdomen to confirm the anti-adhesion effects. Tissue regeneration and inflammatory responses at the injury site were further analyzed using hematoxylin and eosin staining, Masson's trichrome staining, and Ki-67 immunohistochemistry. The ECM-based hemostatic agent significantly reduced the bleeding time compared to conventional products and markedly reduced adhesion formation. In the experimental group, the agent enhanced cell attachment and proliferation at the damaged tissue site, to facilitate the natural tissue regeneration process, without inducing inflammatory or pathological changes. The developed composite hemostatic agent could overcome the limitations of existing products by integrating three crucial functions: rapid hemostasis, preventing adhesion, and promoting tissue regeneration. These findings suggest the potential for hepatocyte proliferation and tissue remodeling, which require further validation, and indicate promising applicability in complex surgical environments.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145194061","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}
Lorena Duarte-Peña, Sheila I Peña-Corona, Luis E López-Jácome, Isaac Ignacio Zepeda González, Hernán Cortés, Gerardo Leyva-Gómez
{"title":"Antimicrobial self-healing injectable hydrogels based on chitosan, collagen, and polyvinyl alcohol for chronic wound treatment.","authors":"Lorena Duarte-Peña, Sheila I Peña-Corona, Luis E López-Jácome, Isaac Ignacio Zepeda González, Hernán Cortés, Gerardo Leyva-Gómez","doi":"10.1088/1748-605X/ae079f","DOIUrl":"10.1088/1748-605X/ae079f","url":null,"abstract":"<p><p>Chronic wounds stand as a significant challenge to public health due to their high prevalence and complications, such as difficult-to-treat infections. The present study focuses on developing antimicrobial self-healing injectable hydrogels composed of chitosan (CS), collagen (CG), and polyvinyl alcohol (PVA) for the noninvasive treatment of chronic wounds with complex geometries. The hydrogels were synthesized through physical crosslinking via hydrogen bonds and ionic interactions, achieved through the freeze-thaw method and pH variations, resulting in materials with dynamic bonds. This feature endowed hydrogels with self-healing capability, allowing injection, adaptation to wound shapes, and recovery of properties after application. The hydrogels exhibited a vapor transmission rate of around 2500-3500 g m<sup>-2</sup>d<sup>-1</sup>, a pH range of 5.2-6.2, 40%-110% swelling, and degradation occurring within 4-48 h, which are within ranges known to support wound regeneration. Rheological analysis revealed viscoelastic and pseudoplastic behavior, and a self-healing capacity of up to 83% after deformation. Hydrogels also presented injection forces below 40 N, ensuring ease of handling. Additionally, hydrogels presented suitable blood compatibility and strong antimicrobial properties, achieving over 99% inhibition against microorganisms commonly associated with chronic wounds. Finally, all hydrogels demonstrate low irritability in the primary skin irritation assay, increased skin moisture, and decreased skin temperature, which are features that could support the wound healing process. These results highlight the potential of these materials for chronic wound treatment, offering a unique combination of natural polymer composition, injectability, self-healing, antimicrobial properties, skin-moisturizing effect, and low irritation potential.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145076090","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}
Isaak J Thornton, Kathryn R Zimlich, Matthew W Fields, James N Wilking
{"title":"Characterizing spatiotemporal microbial colony distributions in printed PEG-DA hydrogel films.","authors":"Isaak J Thornton, Kathryn R Zimlich, Matthew W Fields, James N Wilking","doi":"10.1088/1748-605X/ae066e","DOIUrl":"10.1088/1748-605X/ae066e","url":null,"abstract":"<p><p>Biofilms are surface-attached microbial communities that play vital roles in natural ecosystems and contribute to persistent problems in medicine and industry. These communities exhibit heterogeneous chemical, physical, and physiological properties, which are governed by reciprocal structure-function relationships. Linking structure to function is crucial for understanding biofilm physiology but remains challenging due to the structural complexity of naturally formed biofilms. Bioprinting offers exquisite control over biofilm structure and holds potential for systematically exploring these relationships; however, the microscale colony distributions that emerge within hydrogel-based print resins remain unexplored. To address this, we use light-based bioprinting to create single-layer hydrogel films containing homogeneously dispersed<i>Pseudomonas fluorescens</i>bacteria and characterize the spatiotemporal distribution of colonies that develop within these films. We systematically vary the concentration of bacteria over nearly three orders of magnitude, track colony growth using microscopy, and quantify structural features with image analysis. We observe empirical relationships between initial cell concentration and key structural features: colony size, colony volume, total biovolume, and characteristic gradient length scale. This knowledge can be used to print microbial communities with well-defined features, is readily applicable to more complex three-dimensional shapes, and provides a tool for advancing our understanding of microbial communities.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145055539","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}
{"title":"Biocompatible hydrogel microspheres based on modified silk fibroin and gelatin for injectable 3D bone tissue scaffolds.","authors":"Changsheng Lu, Runqing Shen, Xiao Wang","doi":"10.1088/1748-605X/ae084c","DOIUrl":"10.1088/1748-605X/ae084c","url":null,"abstract":"<p><p>Currently investigated two-dimensional cell culture systems are typically inadequate for large-scale cell expansion and prone to causing altered cell morphology, aberrant differentiation, and distorted protein expression. To overcome these limitations, a glycidyl methacrylate-modified silk fibroin (SFMA)/methacrylic anhydride-modified gelatin (GelMA) interpenetrating polymer network hydrogel (SFMA-GelMA) was developed via microfluidic fabrication for three-dimensional (3D) bone tissue engineering applications. With increased SFMA content, the molecular chains in SFMA-GelMA undergo a structural transformation from random coil to<i>β</i>-sheet and<i>β</i>-crystallite, enhancing storage modulus to about 500 Pa and extending degradation duration from about 47.7% to 84.3% mass retention over 7d. The higher GelMA content with the arginine-glycine-aspartic acid sequence in SFMA-GelMA facilitated early cell adhesion, provided interconnected pores (5-80 μm diameter), and promoted the osteogenic differentiation of MC3T3-E1preosteoblasts in 3D culture, as confirmed by alkaline phosphatase activity up to about 45 U mg<sup>-1</sup>protein. Overall, SFMA-GelMA shows substantial potential as a 3D cell culture scaffold and injectable material for regenerative medicine, particularly in bone tissue engineering.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145081833","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}
Yong Sun, Yizhi Zhang, Ziyan Wei, Xuezhou Liu, Zhaoxi Wang, Kun Wang, Xuewen Kang
{"title":"Black phosphorus nanosheets in orthopedics: from material fabrications to therapeutic prospects.","authors":"Yong Sun, Yizhi Zhang, Ziyan Wei, Xuezhou Liu, Zhaoxi Wang, Kun Wang, Xuewen Kang","doi":"10.1088/1748-605X/ae0548","DOIUrl":"10.1088/1748-605X/ae0548","url":null,"abstract":"<p><p>In recent years, the incidence of orthopedic diseases has increased significantly, while traditional treatments often face limitations such as limited efficacy and pronounced side effects. The development of nanomedicine technology provides novel strategies for orthopedic disease treatment. As an emerging two-dimensional nanomaterial, black phosphorus nanosheets (BPNSs) demonstrate remarkable potential in the treatment of orthopedic diseases due to their unique physicochemical properties, superior biocompatibility, and the fact that their degradation product-elemental phosphorus-constitutes an essential component of bone tissue. This review systematically summarizes the fundamental properties of BPNS, their preparation methods (mechanical exfoliation, chemical vapor deposition, liquid exfoliation, and electrochemical exfoliation), and functional modification strategies (surface covalent coupling, ion loading, and surface coating). We then focus on analyzing research progress in multiple clinical orthopedic applications including bone regeneration, bone defect repair, treatment of degenerative bone diseases, bone tumor therapy, wound healing promotion and orthopedic image-guided applications. Simultaneously, this review objectively discusses key challenges facing clinical translation of BPNS, including long-term biosafety concerns, large-scale preparation technology limitations, and insufficient mechanistic studies, while proposing future research directions. We believe that with further advancements in materials science, nanotechnology, and biomedical engineering, BPNS will become a novel nanomedicine in orthopedic treatment, offering patients more effective and safer therapeutic options.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145031221","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}
Mohd Yaqub Khan, Jen-Kun Chen, Lokesh Agrawal, Garima Joshi, Yu-Ting Chuang, Cheng-An J Lin, Min-Hua Chen
{"title":"Shortwave infrared imaging-guided radiosensitization using rare-earth-doped nanoparticles delivered via microneedles for enhanced melanoma therapy.","authors":"Mohd Yaqub Khan, Jen-Kun Chen, Lokesh Agrawal, Garima Joshi, Yu-Ting Chuang, Cheng-An J Lin, Min-Hua Chen","doi":"10.1088/1748-605X/ae066f","DOIUrl":"10.1088/1748-605X/ae066f","url":null,"abstract":"<p><p>Melanoma remains a major global health challenge due to the uncontrolled growth of abnormal skin cells, resistance to conventional therapies, and poor prognosis in advanced cases. Localized, early-stage melanoma, defined as melanoma confined to the skin without regional or distant spread, offers a critical treatment window, as thin lesions are often curable with surgical excision. However, delays in treatment allow progression to lymph node involvement and distant metastasis, which worsen prognosis and limit available therapies. Although surgery and radiotherapy remain standard options, they often struggle with limitations like incomplete melanoma targeting, damage to healthy tissues, and treatment resistance. To address these challenges, we explored a more precise radiotherapy approach aimed at enhancing treatment efficacy while minimizing harm to surrounding tissues. In this study, we investigated the potential of rare-earth-doped nanoparticles (RENPs) as radiosensitizers by integrating them with microneedles (MNs) and shortwave infrared (SWIR) imaging to improve the precision of radiotherapy for localized, early-stage melanoma treatment. RENPs were synthesized using a modified thermal decomposition method and surface-modified them with Tween 20 (Tw) to facilitate their transition into the aqueous phase for biological applications. Incorporating RENP-Tw into MNs enabled precise and localized delivery into melanoma tissue. Meanwhile SWIR imaging, with its deep tissue penetration and high contrast resolution, allowed real-time monitoring of RENP-Tw localization, ensuring optimal radiosensitization at the melanoma site. Our<i>in vivo</i>studies demonstrated that RENP-Tw/MNs significantly enhanced radiation-induced cell death in melanoma-bearing mice while minimizing systemic toxicity. Moreover, SWIR imaging revealed sustained luminescence of RENP-Tw/MNs at the melanoma site, further supporting precise radiotherapy with improved therapeutic outcomes. This innovative approach addresses the limitations of conventional radiotherapy by improving melanoma specificity, reducing off-target effects, and enhancing radiosensitization efficiency. Overall, our findings suggest that RENP-Tw/MNs hold potential as an effective strategy for advancing localized, early-stage melanoma treatment through precise, imaging-guided radiotherapy.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145056231","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}
{"title":"TAPP@NBs combined with GSNO to enhance the anti-liver cancer effect of sonodynamic therapy.","authors":"Chunyue Wang, Wang Xiaodong, Fengjiao Chen, Huimin Tian, Yichi Chen, Bolin Wu, Wen Cheng","doi":"10.1088/1748-605X/ae0c4e","DOIUrl":"https://doi.org/10.1088/1748-605X/ae0c4e","url":null,"abstract":"<p><strong>Objective: </strong>Due to its significant tissue penetration, sonodynamic therapy (SDT) is rapidly becoming a viable alternative to traditional photodynamic therapy. However, the therapeutic efficacy of a single SDT treatment is constrained by the prolonged hypoxia of the tumor, rendering it ineffective for disease cure. This study employed SDT in conjunction with nitric oxide (NO) gas to induce apoptosis and ferroptosis in hepatocellular carcinoma (HCC) cells, aiming to facilitate cancer treatment.
Approach: In this work, the synthesis of TAPP@NBs involved the encapsulation of 5,10,15,20-Tetrakis (4-aminophenyl) porphyrin within a hydrated phospholipid film. Structural and morphological properties of the nanoparticles were analyzed using dynamic light scattering and transmission electron microscopy. In vitro, the anticancer cytotoxicity of TAPP@NBs was examined using HepG2 and HUH7 hepatoma cell lines. Identification of TAPP@NBs mediated SDT for HCC therapy through the mitochondrial pathway was achieved by staining with 2, 7-dichlorodihydrofluorescein diacetate (DCFH-DA) and 5,5', 6,6'-tetrachloro-1, 1', 3,3'-tetraethylimidacloprid iodide (JC-1). Quantification of cell viability and apoptosis was conducted using CCK-8 and flow cytometry. Evidence of iron death was established using the glutathione (GSH) metabolism assay and malondialdehyde (MDA) assay. The cellular migratory capacity was assessed using a transwell assay.
Main results: TAPP@NBs could effectively generate deadly amounts of reactive oxygen species (ROS) through ultrasonic irradiation. Under ultrasonography, nitrosoglutathione (GSNO) could serve as a NO donor and contribute to gas treatment, so significantly improving the effectiveness of SDT. Significant reductions in HCC cell activity and migration ability were observed after the combination of SDT with NO gas therapy.
Significance: SDT in conjunction with NO gas therapy can effectively induce apoptosis and ferroptosis in HCC cells. This study presents a novel approach for the optimization of tumor treatment.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145180538","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}
Peng Chen, Jiawei Lu, Yi Liu, Hongwei Wang, Yaguang Han, Xiaoji Luo
{"title":"Stem cell membrane-coated rough mesoporous silica nanoparticles for enhanced osteogenic differentiation and bone repair via dexamethasone delivery.","authors":"Peng Chen, Jiawei Lu, Yi Liu, Hongwei Wang, Yaguang Han, Xiaoji Luo","doi":"10.1088/1748-605X/ae0bda","DOIUrl":"https://doi.org/10.1088/1748-605X/ae0bda","url":null,"abstract":"<p><p>Mesoporous silica nanoparticles (MSNs) have been demonstrated to promote osteoblast differentiation; however, the unclear impact of their surface roughness on osteogenesis, coupled with inadequate targeting capability and suboptimal therapeutic outcomes, presents major challenges. Herein, we developed a biomimetic nanoplatform, CM@DEX-R-MSN, by coating dexamethasone (DEX) loaded-rough MSN (R-MSN) with mesenchymal stem cell (MSC) membranes (CM) to enhance osteogenic differentiation of MSCs for improved bone regeneration. The CM@DEX-R-MSN showed retained rough surfaces with a hydrodynamic diameter of 164.35 ± 5.81 nm, a Zeta potential of -11.98 ± 1.37 mV with good MSC membrane integrity, negligible cytotoxicity both in vitro and in vivo. CM@DEX-R-MSN exhibited significantly enhanced MSC internalization compared to uncoated MSN. They markedly upregulated alkaline phosphatase activity, osteogenic markers, and mineralization nodule formation in vitro. In bone defect model established in rabbits, CM@DEX-R-MSN restored bone volume and prolonged retention at the defect site. More importantly, we experimentally observed that both R-MSN and CM-coated nanoparticles exhibited superior osteogenic differentiation effects compared to conventional MSNs and non-coated counterparts, respectively-with CM@DEX-R-MSN demonstrating the most potent efficacy. Our results demonstrated that CM@DEX-R-MSN synergistically integrates MSC membrane-mediated homotypic targeting, nanotopography of R-MSN, and DEX-driven osteogenic differentiation, offering a promising targeted therapeutic strategy for bone regeneration. Their enhanced biocompatibility, osteogenic efficacy, and sustained retention underscore its translational potential for orthopedic applications.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145151956","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}
{"title":"Glucocorticoid receptor-mediated delivery of paclitaxel and anticancer gene p53 for oral cancer therapeutics.","authors":"Kalyani Sakhare, Dwaipayan Bhattacharya, Chhavi Dhiman, Priyanka Erukulla, Srija Bhattacharya, Aasia Ansari, Piyush Khandelia, Anjaneyulu Eanti, Rajkumar Banerjee, Kumar Pranav Narayan","doi":"10.1088/1748-605X/ae05a3","DOIUrl":"10.1088/1748-605X/ae05a3","url":null,"abstract":"<p><p>Targeting glucocorticoid Receptors (GR) induces gluconeogenesis in cancer cells, potentially disrupting their glycolytic dependency and acidic tumor microenvironment (TME), thereby creating an energetically unfavourable state and reducing drug resistance by impairing the acid reflux mechanism. Based on this rationale, we developed a GR-mediated liposomal co-delivery system, D1XP-p53, carrying the tumor suppressor gene, p53, and the chemotherapeutic drug, paclitaxel, to overcome the limitations of conventional anti-cancer therapies and to assess whether wild-type p53 enhances the anti-cancer activity of paclitaxel against Oral Squamous Cell Carcinoma (OSCC).<i>In vitro</i>studies demonstrated that D1XP-p53 selectively decreased the viability of OSCC cells and significantly inhibited their migration, invasion, and proliferation. Mechanistic investigations revealed an upregulation of the BAX/BCL2 ratio when oral cancer cells were treated with D1XP-p53, indicating the activation of intrinsic apoptotic pathways. The efficacy of D1XP-p53 was further validated in 3D spheroid models using MOC2 and FaDu cell lines, where it significantly reduced spheroid-forming ability and upregulated E-cadherin expression, indicating its potential role in enhancing anti-cancer activity and mitigating cellular migration.<i>In vivo</i>experiments using a murine model of OSCC with MOC2 cells showed a marked reduction in tumor volume in mice treated with D1XP-p53, with minimal systemic toxicity as assessed by H&E staining and biodistribution analysis. Considering the crucial role of TME components such as tumor-associated macrophages, cancer stem cells, and growth factors in tumor progression and metastasis, we further evaluated the impact of our delivery system, D1XP-p53, on these elements. We observed that D1XP-p53 treatment in mice significantly upregulated the M1/M2 ratios and decreased the<i>c-myc</i>and<i>SOX2</i>expression, indicating the potential role of the delivery system in modulating the TME components. These findings collectively demonstrate that the GR-targeted co-delivery system, D1XP-p53, enhances anti-cancer activity and modulates the TME, offering a promising multi-modal treatment against aggressive oral cancer.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145034800","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}
{"title":"Bilayer oxidized sodium alginate-carboxymethyl chitosan hydrogel microspheres enable sustained BMP-2 release for enhanced bone regeneration.","authors":"Yafei Yuan, Xige Zhao, Jiangqi Hu, Yixuan Zhu, Xuening Deng, Qingsong Jiang","doi":"10.1088/1748-605X/ae0778","DOIUrl":"https://doi.org/10.1088/1748-605X/ae0778","url":null,"abstract":"<p><p>Bone morphogenetic protein-2 (BMP-2) is a highly potent osteoinductive factor that has received approval from the U.S. Food and Drug Administration (FDA) due to its significant osteogenic properties. Nonetheless, its clinical utility is limited by adverse effects linked to supraphysiological dosing and its brief half-life. Consequently, there is a pressing need for a safe and effective delivery system to enable the sustained release of BMP-2. In this study, we have developed bilayer-structured oxidized sodium alginate-carboxymethyl chitosan (OAC) microspheres through the application of electrospraying and the Schiff reaction. The inner layer, composed of oxidized sodium alginate, electrostatically adsorbs BMP-2, while the porous polyelectrolyte membrane on the surface enhances adsorption, thereby effectively regulating the prolonged and controlled release of BMP-2. We assessed the minimal osteogenic induction concentration of BMP-2 on rat bone marrow mesenchymal stem cells (rBMSCs) to optimize the BMP-2 loading concentration within the microspheres. In vitro experiments demonstrated that the bilayer membrane structure of the hydrogel microspheres significantly delayed the release of BMP-2, facilitating a long-term, sustained release. Furthermore, the microspheres facilitated the proliferation, migration, and osteogenic differentiation of rat bone marrow-derived mesenchymal stem cells (rBMSCs). The osteogenic-promoting efficacy of the BMP-2-encapsulated OAC microspheres was further corroborated in vivo through implantation alongside calcium phosphate cement into the dorsal region of nude mice. Collectively, the BMP-2encapsulated OAC microspheres we developed constitute a promising clinical approach to augment scaffold degradation and osteogenesis for the repair of bone defects.</p>","PeriodicalId":72389,"journal":{"name":"Biomedical materials (Bristol, England)","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145076149","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}