Pub Date : 2026-02-01Epub Date: 2025-08-06DOI: 10.1016/j.biomaterials.2025.123599
Lin Zhu, Mali Zu, Feifan Wu, Xu Ma, Shuhui Zhang, Tianchen Zhang, Xingfan Li, Huiru Yang, Zhenqi Li, Zetao Yu, Ruijie Nie, Tianjiao Ji, Xiuping Zhang, Rong Liu
Portal vein tumor thrombus (PVTT) is a common and severe indicator in advanced hepatocellular carcinoma (HCC), characterized by a poor prognosis and limited response to existing therapies. Cancer-associated fibroblasts (CAFs) play an important role in promoting HCC metastasis and contribute to resistance against sorafenib (SOR) resistance, which is a standard treatment for advanced HCC. The data from single-cell RNA sequencing highlights the critical role of C-X-C motif chemokine ligand 12 (CXCL12) in the activation of CAFs. To address these challenges, we develop a PVTT-targeted nanocarrier designed to co-deliver small interfering RNA (siRNA) and a multikinase inhibitor, aiming to enhance therapeutic outcomes for PVTT. This novel lipid-coated polylactide-co-glycolide nanoparticle system effectively downregulate CXCL12 expression in CAFs, leading to their inactivation and subsequent reshaping of the tumor microenvironment. The resulting modulation of the tumor microenvironment significantly suppress tumor cell migration, invasion, and resistance to SOR, thereby demonstrating potent anti-tumor effects in orthotopic mouse models of PVTT. Furthermore, RNA sequencing reveals key regulatory pathways and genes associated with the inhibition of SOR resistance and PVTT formation mediated by these nanoparticles. These findings suggest that modulating the tumor microenvironment, combined with targeted anti-tumor therapies, offers a promising strategy for treating HCC patients with PVTT.
{"title":"Cancer-associated fibroblasts regulating nanomedicine to overcome sorafenib resistance in hepatocellular carcinoma with portal vein tumor thrombus.","authors":"Lin Zhu, Mali Zu, Feifan Wu, Xu Ma, Shuhui Zhang, Tianchen Zhang, Xingfan Li, Huiru Yang, Zhenqi Li, Zetao Yu, Ruijie Nie, Tianjiao Ji, Xiuping Zhang, Rong Liu","doi":"10.1016/j.biomaterials.2025.123599","DOIUrl":"10.1016/j.biomaterials.2025.123599","url":null,"abstract":"<p><p>Portal vein tumor thrombus (PVTT) is a common and severe indicator in advanced hepatocellular carcinoma (HCC), characterized by a poor prognosis and limited response to existing therapies. Cancer-associated fibroblasts (CAFs) play an important role in promoting HCC metastasis and contribute to resistance against sorafenib (SOR) resistance, which is a standard treatment for advanced HCC. The data from single-cell RNA sequencing highlights the critical role of C-X-C motif chemokine ligand 12 (CXCL12) in the activation of CAFs. To address these challenges, we develop a PVTT-targeted nanocarrier designed to co-deliver small interfering RNA (siRNA) and a multikinase inhibitor, aiming to enhance therapeutic outcomes for PVTT. This novel lipid-coated polylactide-co-glycolide nanoparticle system effectively downregulate CXCL12 expression in CAFs, leading to their inactivation and subsequent reshaping of the tumor microenvironment. The resulting modulation of the tumor microenvironment significantly suppress tumor cell migration, invasion, and resistance to SOR, thereby demonstrating potent anti-tumor effects in orthotopic mouse models of PVTT. Furthermore, RNA sequencing reveals key regulatory pathways and genes associated with the inhibition of SOR resistance and PVTT formation mediated by these nanoparticles. These findings suggest that modulating the tumor microenvironment, combined with targeted anti-tumor therapies, offers a promising strategy for treating HCC patients with PVTT.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"325 ","pages":"123599"},"PeriodicalIF":12.9,"publicationDate":"2026-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144797756","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-02-01Epub Date: 2025-08-06DOI: 10.1016/j.biomaterials.2025.123604
Xuan Li, Xinxin Luo, Ye He, Bikun Zhou, Kun Xu, Qian Huang, Xiao Jiang, Hongwei Xiong, Xuezhe Liu, Shaopeng Liu, Bailong Tao, Peng Liu, Kaiyong Cai
The senescence of mesenchymal stem cells (MSCs) leads to the significant change of their metabolic activity and physiological behaviors. In the context of orthopedic treatment, the osteointegration of titanium implant is largely affected by MSC aging, imposing considerable limitations on its long-term application. In this study, a surface modification on titanium implants was designed to enhance osteointegration by effectively regulating the functions of senescent MSC: A typical micro-nano topological structure was established on the implant surface to improve the osteogenic differentiation of MSCs. Then a functional hydrogel coating was covalently modified to the implant surface through a poly-dopamine layer. For senescent MSCs, firstly, the coating can eliminate the activation of senescence-associated secretory phenotype (SASP) of senescent MSCs by micro-nano topological structure, and it accelerated the proliferation of non-senescent MSCs by the reactive oxygen species (ROS) scavenging. With the degradation of the hydrogel coating, the composition of stem cell pool around the implant interfaces gradually rejuvenated, as the number of non-senescent MSCs increased and senescent MSCs decreased. Meanwhile, the exposed micro-nano topological structure showed significant effect on the osteogenic differentiation of MSCs, and ultimately promoted the osteointegration in aging rats. These results provided promising insights for the design and application of orthopedic titanium implants for aging patients.
{"title":"Enhanced osteointegration of implants in aged rats via a stem cell pool aging reversion strategy.","authors":"Xuan Li, Xinxin Luo, Ye He, Bikun Zhou, Kun Xu, Qian Huang, Xiao Jiang, Hongwei Xiong, Xuezhe Liu, Shaopeng Liu, Bailong Tao, Peng Liu, Kaiyong Cai","doi":"10.1016/j.biomaterials.2025.123604","DOIUrl":"10.1016/j.biomaterials.2025.123604","url":null,"abstract":"<p><p>The senescence of mesenchymal stem cells (MSCs) leads to the significant change of their metabolic activity and physiological behaviors. In the context of orthopedic treatment, the osteointegration of titanium implant is largely affected by MSC aging, imposing considerable limitations on its long-term application. In this study, a surface modification on titanium implants was designed to enhance osteointegration by effectively regulating the functions of senescent MSC: A typical micro-nano topological structure was established on the implant surface to improve the osteogenic differentiation of MSCs. Then a functional hydrogel coating was covalently modified to the implant surface through a poly-dopamine layer. For senescent MSCs, firstly, the coating can eliminate the activation of senescence-associated secretory phenotype (SASP) of senescent MSCs by micro-nano topological structure, and it accelerated the proliferation of non-senescent MSCs by the reactive oxygen species (ROS) scavenging. With the degradation of the hydrogel coating, the composition of stem cell pool around the implant interfaces gradually rejuvenated, as the number of non-senescent MSCs increased and senescent MSCs decreased. Meanwhile, the exposed micro-nano topological structure showed significant effect on the osteogenic differentiation of MSCs, and ultimately promoted the osteointegration in aging rats. These results provided promising insights for the design and application of orthopedic titanium implants for aging patients.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"325 ","pages":"123604"},"PeriodicalIF":12.9,"publicationDate":"2026-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144803048","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-02-01Epub Date: 2025-08-05DOI: 10.1016/j.biomaterials.2025.123602
Ming Li, Shengzhe Zhou, Qiang Yu, Chenxi Wang, Haoyi Chen, Yingying Ma, Huizhen Fan, Tao Ni, Min Lu, Min Yao
Chronic diabetic wounds are characterized by hypoxia, persistent microbial infection, and impaired healing, posing significant challenges to conventional therapies. Herein, we present a novel sprayable double-network hydrogel platform designed to achieve efficient antimicrobial activity and accelerated wound repair under hypoxic conditions by leveraging a type I photodynamic therapy (PDT) and immune-metabolic regulatory strategy. Specifically, we employ salvianolic acid B (SAB) to form a self-assembled hydrogel (SAB-gel) and incorporate fibrin to construct a robust and acidic double-network SAB/F-gel with enhanced mechanical strength and acidic environment. Concurrently, thymoquinone (TQ) and chlorin e6 (Ce6) are self-assembled via hydrophobic interactions to form TQ/Ce6 nanoparticles (TQ/Ce6 NPs) and embedded in the SAB/F-gel, to fabricate the TQ/Ce6@SAB/F-gel. Under low-oxygen conditions, TQ acts as an electron-transfer mediator, enabling Ce6 to generate abundant superoxide anions (·O2-) via type I PDT under red light (RL) irradiation. These ·O2- are subsequently converted into hydrogen peroxide (H2O2) and hydroxyl radicals (·OH) in the acidic environment provided by acidic SAB/F-gel, thereby reducing the dependence on oxygen and maintaining potent antimicrobial efficacy against MRSA, Pseudomonas aeruginosa (Pa), Acinetobacter baumannii (Ab), Escherichia coli (E. coli) and Candida albicans (Ca). In vitro experiments demonstrated that TQ/Ce6@SAB/F-gel regulates macrophage M2 polarization and promotes endothelial cell proliferation, migration, and tube formation via the immune-metabolic regulatory pathways. When applied to MRSA-infected diabetic wounds in mice, the hydrogel in combination with RL completely eradicated bacteria, promoted collagen deposition and angiogenesis, and significantly accelerated wound closure, as demonstrated by histological examination and transcriptome sequencing. This work offers a versatile, biocompatible, and oxygen-independent PDT-based hydrogel system for the treatment of refractory infected diabetic wounds, offering potential for clinical translation and improved patient outcomes.
慢性糖尿病伤口以缺氧、持续微生物感染和愈合受损为特征,对传统治疗提出了重大挑战。在此,我们提出了一种新型的可喷雾双网络水凝胶平台,旨在利用I型光动力疗法(PDT)和免疫代谢调节策略,在缺氧条件下实现有效的抗菌活性和加速伤口修复。具体而言,我们利用丹酚酸B (SAB)形成自组装水凝胶(SAB-gel),并加入纤维蛋白构建坚固的酸性双网络SAB/ f-凝胶,增强了机械强度和酸性环境。同时,百里醌(TQ)和氯e6 (Ce6)通过疏水相互作用自组装形成TQ/Ce6纳米颗粒(TQ/Ce6 NPs)并嵌入SAB/ f-凝胶中,制备TQ/Ce6@SAB/ f-凝胶。在低氧条件下,TQ作为电子转移介质,使Ce6在红光(RL)照射下通过I型PDT产生丰富的超氧阴离子(·O2-)。这些·O2-随后在酸性SAB/ f -凝胶提供的酸性环境中转化为过氧化氢(H2O2)和羟基自由基(·OH),从而降低对氧的依赖,并保持对MRSA、铜绿假单胞菌(Pa)、鲍曼不动杆菌(Ab)、大肠杆菌(E. coli)和白色念珠菌(Ca)的有效抗菌效果。体外实验表明,TQ/Ce6@SAB/F-gel通过免疫代谢调控途径调控巨噬细胞M2极化,促进内皮细胞增殖、迁移和成管。组织学检查和转录组测序结果显示,水凝胶与RL联合应用于mrsa感染的小鼠糖尿病创面,可彻底根除细菌,促进胶原沉积和血管生成,显著加速创面愈合。这项工作为治疗难治性糖尿病感染伤口提供了一种通用的、生物相容性的、不依赖氧的基于pdp的水凝胶系统,为临床转化和改善患者预后提供了潜力。
{"title":"A sprayable TQ/Ce6@SAB/F-gel for accelerating wound healing via hypoxia-tolerant photodynamic therapy and immune-metabolic pathway.","authors":"Ming Li, Shengzhe Zhou, Qiang Yu, Chenxi Wang, Haoyi Chen, Yingying Ma, Huizhen Fan, Tao Ni, Min Lu, Min Yao","doi":"10.1016/j.biomaterials.2025.123602","DOIUrl":"10.1016/j.biomaterials.2025.123602","url":null,"abstract":"<p><p>Chronic diabetic wounds are characterized by hypoxia, persistent microbial infection, and impaired healing, posing significant challenges to conventional therapies. Herein, we present a novel sprayable double-network hydrogel platform designed to achieve efficient antimicrobial activity and accelerated wound repair under hypoxic conditions by leveraging a type I photodynamic therapy (PDT) and immune-metabolic regulatory strategy. Specifically, we employ salvianolic acid B (SAB) to form a self-assembled hydrogel (SAB-gel) and incorporate fibrin to construct a robust and acidic double-network SAB/F-gel with enhanced mechanical strength and acidic environment. Concurrently, thymoquinone (TQ) and chlorin e6 (Ce6) are self-assembled via hydrophobic interactions to form TQ/Ce6 nanoparticles (TQ/Ce6 NPs) and embedded in the SAB/F-gel, to fabricate the TQ/Ce6@SAB/F-gel. Under low-oxygen conditions, TQ acts as an electron-transfer mediator, enabling Ce6 to generate abundant superoxide anions (·O<sub>2</sub><sup>-</sup>) via type I PDT under red light (RL) irradiation. These ·O<sub>2</sub><sup>-</sup> are subsequently converted into hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>) and hydroxyl radicals (·OH) in the acidic environment provided by acidic SAB/F-gel, thereby reducing the dependence on oxygen and maintaining potent antimicrobial efficacy against MRSA, Pseudomonas aeruginosa (Pa), Acinetobacter baumannii (Ab), Escherichia coli (E. coli) and Candida albicans (Ca). In vitro experiments demonstrated that TQ/Ce6@SAB/F-gel regulates macrophage M2 polarization and promotes endothelial cell proliferation, migration, and tube formation via the immune-metabolic regulatory pathways. When applied to MRSA-infected diabetic wounds in mice, the hydrogel in combination with RL completely eradicated bacteria, promoted collagen deposition and angiogenesis, and significantly accelerated wound closure, as demonstrated by histological examination and transcriptome sequencing. This work offers a versatile, biocompatible, and oxygen-independent PDT-based hydrogel system for the treatment of refractory infected diabetic wounds, offering potential for clinical translation and improved patient outcomes.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"325 ","pages":"123602"},"PeriodicalIF":12.9,"publicationDate":"2026-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144811478","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-02-01Epub Date: 2025-08-05DOI: 10.1016/j.biomaterials.2025.123595
Yixin Liu, Xinjian Yang, Ya Miao, Taoping Chen, Wenyan Gao, Guoqiang Zhou, Guang Jia, Xiaosong Yang, Jinchao Zhang, Yi Jin
Osteoarthritis (OA), a prevalent degenerative joint disease, currently lacks effective therapeutic options beyond symptomatic relief. Persistent inflammation and impaired cartilage repair accelerate the disease progression. The enzyme inducible nitric oxide synthase (iNOS) contributes to OA by producing nitric oxide (NO), which intensifies inflammation and inhibits cartilage regeneration. Traditional iNOS inhibitors have demonstrated limited efficacy due to inadequate targeted release and uncoordinated control over inflammation. In this study, we developed a self-supported DNAzyme-based DNA hydrogel using rolling circle amplification (RCA) technology to deliver iNOS-targeting DNAzymes and bone marrow mesenchymal stem cell-derived exosomes (BMSC-exos) in response to inflammation. The hydrogel incorporates triglycerol monostearate nanoparticles (TGMS NPs), which degrade under high matrix metalloproteinase (MMP) levels in OA joints, thereby triggering the release of the DNAzymes and exosomes with precision. This targeted delivery modulates the inflammatory microenvironment by reducing pro-inflammatory NO production and supports chondrogenesis by promoting M2 macrophage polarization. In vitro and in vivo analyses reveal that the hydrogel significantly reduces inflammatory cytokine levels, enhances chondrocyte proliferation, and restores extracellular matrix integrity, ultimately slowing OA progression. This smart hydrogel offers a promising ambidextrous strategy for microenvironment modulation and cartilage regeneration, potentially advancing OA treatment.
骨关节炎(OA)是一种常见的退行性关节疾病,目前除了症状缓解之外缺乏有效的治疗选择。持续的炎症和受损的软骨修复加速了疾病的进展。酶诱导型一氧化氮合酶(iNOS)通过产生一氧化氮(NO)促进OA,从而加剧炎症并抑制软骨再生。传统的iNOS抑制剂由于不充分的靶向释放和对炎症的不协调控制而显示出有限的疗效。在这项研究中,我们开发了一种基于dnazyme的自我支持的DNA水凝胶,使用rolling circle amplification (RCA)技术来递送靶向inos的DNAzymes和骨髓间充质干细胞衍生的外泌体(BMSC-exos),以应对炎症。该水凝胶含有甘油三酯单硬脂酸纳米颗粒(TGMS NPs),该纳米颗粒在OA关节的高基质金属蛋白酶(MMP)水平下降解,从而精确触发DNAzymes和外泌体的释放。这种靶向递送通过减少促炎NO的产生来调节炎症微环境,并通过促进M2巨噬细胞极化来支持软骨形成。体外和体内分析表明,水凝胶可显著降低炎症细胞因子水平,增强软骨细胞增殖,恢复细胞外基质完整性,最终减缓OA进展。这种智能水凝胶为微环境调节和软骨再生提供了一种有前途的双灵巧策略,有可能推进OA治疗。
{"title":"Self-supported DNA hydrogel facilitates microenvironment remodeling and cartilage repair to prevent osteoarthritis progression via an ambidextrous strategy.","authors":"Yixin Liu, Xinjian Yang, Ya Miao, Taoping Chen, Wenyan Gao, Guoqiang Zhou, Guang Jia, Xiaosong Yang, Jinchao Zhang, Yi Jin","doi":"10.1016/j.biomaterials.2025.123595","DOIUrl":"10.1016/j.biomaterials.2025.123595","url":null,"abstract":"<p><p>Osteoarthritis (OA), a prevalent degenerative joint disease, currently lacks effective therapeutic options beyond symptomatic relief. Persistent inflammation and impaired cartilage repair accelerate the disease progression. The enzyme inducible nitric oxide synthase (iNOS) contributes to OA by producing nitric oxide (NO), which intensifies inflammation and inhibits cartilage regeneration. Traditional iNOS inhibitors have demonstrated limited efficacy due to inadequate targeted release and uncoordinated control over inflammation. In this study, we developed a self-supported DNAzyme-based DNA hydrogel using rolling circle amplification (RCA) technology to deliver iNOS-targeting DNAzymes and bone marrow mesenchymal stem cell-derived exosomes (BMSC-exos) in response to inflammation. The hydrogel incorporates triglycerol monostearate nanoparticles (TGMS NPs), which degrade under high matrix metalloproteinase (MMP) levels in OA joints, thereby triggering the release of the DNAzymes and exosomes with precision. This targeted delivery modulates the inflammatory microenvironment by reducing pro-inflammatory NO production and supports chondrogenesis by promoting M2 macrophage polarization. In vitro and in vivo analyses reveal that the hydrogel significantly reduces inflammatory cytokine levels, enhances chondrocyte proliferation, and restores extracellular matrix integrity, ultimately slowing OA progression. This smart hydrogel offers a promising ambidextrous strategy for microenvironment modulation and cartilage regeneration, potentially advancing OA treatment.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"325 ","pages":"123595"},"PeriodicalIF":12.9,"publicationDate":"2026-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144803049","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Although autografts and allografts remain common for bone defect repair, they entail donor-site morbidity, limited availability, and potential immune rejection. The development of tissue engineering has provided a potential solution to overcome these and facilitate effective bone regeneration. Extensive research has confirmed the osteogenic potential of bioactive molecules like Atorvastatin (ATV) and Icariin (ICA). But despite the increasing body of evidence supporting their individual merits, few studies have investigated the synergistic integration of these materials in Nanocomposite scaffolds. A novel three-dimensional scaffold composed of polycaprolactone (PCL), carboxymethyl chitosan (CMCs), and nano-hydroxyapatite (nHA), co-loaded with Icariin and Atorvastatin, and fabricated using the freeze-casting technique, is described. This study aimed to evaluate the scaffold's effectiveness in promoting calvarial bone regeneration in Wistar rats, contributing to the advancement of biomaterials in bone tissue engineering. Scaffolds containing PCL/CMCs/nHA with 0.1% ICA and 0.1% ATV were fabricated using the freeze-casting method. In vitro assessments were conducted to evaluate the biomechanical and physiological properties of the scaffolds. In vivo experiments involved implanting the scaffolds into calvarial bone defects in six groups of Wistar rats. After 12 weeks, histological analysis was performed to assess bone regeneration, including fibrous tissue formation, bone formation, osteon development, and osteoblast cell numbers and fibroblast cell numbers. After 72 h of incubation, the PCL/CMCs/nHA/ATO/ICA scaffold significantly enhanced cell viability compared to other groups, however, the differences observed between the other groups were not statistically significant. In vivo, results showed significantly greater bone formation, osteon development, and osteoblast numbers in the PCL/CMCs/nHA/ATO/ICA group than in the negative and other groups. The PCL/CMCs/nHA/ATO/ICA scaffold demonstrated superior bone regeneration outcomes, showing comparable performance to autografts in terms of new bone tissue formation, osteon structure, and 72-h cell viability, suggesting its potential as a viable alternative in bone tissue engineering.
{"title":"Investigation of calvarial bone regeneration in a rat model using three-dimensional polycaprolactone/carboxymethyl chitosan nano composite scaffolds containing hydroxyapatite nanoparticles along with the icariin and atorvastatin synthesized by the freeze-casting method.","authors":"Nadia Sadeghi, Fereshteh Shanei, Abouzar Moradi, Atefeh Shamosi, Sepehr Zamani, Majid Salehi","doi":"10.1177/08853282251369228","DOIUrl":"10.1177/08853282251369228","url":null,"abstract":"<p><p>Although autografts and allografts remain common for bone defect repair, they entail donor-site morbidity, limited availability, and potential immune rejection. The development of tissue engineering has provided a potential solution to overcome these and facilitate effective bone regeneration. Extensive research has confirmed the osteogenic potential of bioactive molecules like Atorvastatin (ATV) and Icariin (ICA). But despite the increasing body of evidence supporting their individual merits, few studies have investigated the synergistic integration of these materials in Nanocomposite scaffolds. A novel three-dimensional scaffold composed of polycaprolactone (PCL), carboxymethyl chitosan (CMCs), and nano-hydroxyapatite (nHA), co-loaded with Icariin and Atorvastatin, and fabricated using the freeze-casting technique, is described. This study aimed to evaluate the scaffold's effectiveness in promoting calvarial bone regeneration in Wistar rats, contributing to the advancement of biomaterials in bone tissue engineering. Scaffolds containing PCL/CMCs/nHA with 0.1% ICA and 0.1% ATV were fabricated using the freeze-casting method. In vitro assessments were conducted to evaluate the biomechanical and physiological properties of the scaffolds. In vivo experiments involved implanting the scaffolds into calvarial bone defects in six groups of Wistar rats. After 12 weeks, histological analysis was performed to assess bone regeneration, including fibrous tissue formation, bone formation, osteon development, and osteoblast cell numbers and fibroblast cell numbers. After 72 h of incubation, the PCL/CMCs/nHA/ATO/ICA scaffold significantly enhanced cell viability compared to other groups, however, the differences observed between the other groups were not statistically significant. In vivo, results showed significantly greater bone formation, osteon development, and osteoblast numbers in the PCL/CMCs/nHA/ATO/ICA group than in the negative and other groups. The PCL/CMCs/nHA/ATO/ICA scaffold demonstrated superior bone regeneration outcomes, showing comparable performance to autografts in terms of new bone tissue formation, osteon structure, and 72-h cell viability, suggesting its potential as a viable alternative in bone tissue engineering.</p>","PeriodicalId":15138,"journal":{"name":"Journal of Biomaterials Applications","volume":" ","pages":"727-742"},"PeriodicalIF":2.5,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144846619","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
One of the disadvantages of traditional ophthalmic formulations is their short residence time in the eye. An in situ gel is recommended as a remedy, as it can be converted into a gel upon contact with the eye and adhere for an extended period. Tamarind seed polysaccharide (TSP) is non thermo-sensitive and possesses the necessary properties to be used as a vehicle for administering medication to the eye. However, the administration of medication into the eyes through TSP based in situ gel has not yet been studied. N-isopropyl acrylamide was grafted onto TSP to make it temperature sensitive. Then, a TSP-based thermo-sensitive in situ gel-forming solution loaded with dorzolamide hydrochloride (2% w/v) was developed and evaluated through in vitro, ex vivo, and in vivo tests. The in situ gel forming solution turns into a gel at 37°C. The safety and efficacy of the formulation were confirmed through an in vivo study on rabbit eyes with induced glaucoma. The findings indicate that the in situ gel significantly reduced intraocular pressure (IOP), with effects comparable to those of marketed eye drops.
{"title":"<i>In vitro</i>, <i>ex vivo</i>, and <i>in vivo</i> evaluation of polysaccharide based thermo-sensitive <i>in situ</i> gel for the treatment of glaucoma.","authors":"Sonali Mandal, Nihar Ranjan Das, Kaushik Mukherjee, Tapan Kumar Giri","doi":"10.1177/08853282251369232","DOIUrl":"10.1177/08853282251369232","url":null,"abstract":"<p><p>One of the disadvantages of traditional ophthalmic formulations is their short residence time in the eye. An <i>in situ</i> gel is recommended as a remedy, as it can be converted into a gel upon contact with the eye and adhere for an extended period. Tamarind seed polysaccharide (TSP) is non thermo-sensitive and possesses the necessary properties to be used as a vehicle for administering medication to the eye. However, the administration of medication into the eyes through TSP based <i>in situ</i> gel has not yet been studied. <i>N</i>-isopropyl acrylamide was grafted onto TSP to make it temperature sensitive. Then, a TSP-based thermo-sensitive <i>in situ</i> gel-forming solution loaded with dorzolamide hydrochloride (2% w/v) was developed and evaluated through <i>in vitro</i>, <i>ex vivo</i>, and <i>in vivo</i> tests. The <i>in situ gel</i> forming solution turns into a gel at 37°C. The safety and efficacy of the formulation were confirmed through an <i>in vivo</i> study on rabbit eyes with induced glaucoma. The findings indicate that the <i>in situ</i> gel significantly reduced intraocular pressure (IOP), with effects comparable to those of marketed eye drops.</p>","PeriodicalId":15138,"journal":{"name":"Journal of Biomaterials Applications","volume":" ","pages":"650-665"},"PeriodicalIF":2.5,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144835191","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01Epub Date: 2025-08-29DOI: 10.1177/08853282251369244
Sida Liao, Zijie Sun, Furong Lin, Jingyu Liang, Longmei Guo, Zixin Deng, Xinyao Zhang, Minghui Zhong, Jiamin Zeng, Bu Long, Jiawei Huang, Wenjin Ji, Lan Lan
Study objectives: We aimed to develop a drug-loaded hydrogel-encapsulated chest drain to improve postoperative comfort and recovery in thoracic surgery patients. Methods: The hydrogel was modified with different ratios of glycerol and alginate, then mixed with varying concentrations of ropivacaine and fixed on a simulated chest drain tube using a mould and calcium chloride solution. The morphology, degradation, and slow-release properties of the hydrogel were assessed to identify the most suitable formulation. A bacteriostatic test was conducted using bacterial smear plates. The new chest drain was then implanted in rats using the seldinger method. Pathological changes were observed with imaging techniques such as chest ultrasound and radiographs, while lung function was assessed to evaluate the analgesic effect. After the animal experiments, hematoxylin and eosin (H&E) and Masson staining were performed on relevant tissues to analyze inflammation, and SOD activity was measured to assess oxidative stress levels. Results: The optimal drug-loaded hydrogel for chest drains contained 2% sodium alginate, 10% glycerol, and ropivacaine concentrations between 0.25% and 0.75%. This formulation showed superior morphological characteristics, degradation, and sustained-release properties. It also exhibited excellent bacteriostatic effects. The low-concentration (0.25%) drug-loaded hydrogel demonstrated better analgesic, anti-inflammatory, and oxidative stress-inhibitory effects in animal studies. Conclusions: The modified ropivacaine-alginate hydrogel-encapsulated chest drain offers a promising local slow-release strategy and may contribute to rapid rehabilitation in thoracic surgery.
{"title":"A novel hydrogel-coated chest drain based on ropivacaine-glycerol-alginate hydrogel with construction and application to postoperative thoracic rehabilitation.","authors":"Sida Liao, Zijie Sun, Furong Lin, Jingyu Liang, Longmei Guo, Zixin Deng, Xinyao Zhang, Minghui Zhong, Jiamin Zeng, Bu Long, Jiawei Huang, Wenjin Ji, Lan Lan","doi":"10.1177/08853282251369244","DOIUrl":"10.1177/08853282251369244","url":null,"abstract":"<p><p><b>Study objectives:</b> We aimed to develop a drug-loaded hydrogel-encapsulated chest drain to improve postoperative comfort and recovery in thoracic surgery patients. <b>Methods:</b> The hydrogel was modified with different ratios of glycerol and alginate, then mixed with varying concentrations of ropivacaine and fixed on a simulated chest drain tube using a mould and calcium chloride solution. The morphology, degradation, and slow-release properties of the hydrogel were assessed to identify the most suitable formulation. A bacteriostatic test was conducted using bacterial smear plates. The new chest drain was then implanted in rats using the seldinger method. Pathological changes were observed with imaging techniques such as chest ultrasound and radiographs, while lung function was assessed to evaluate the analgesic effect. After the animal experiments, hematoxylin and eosin (H&E) and Masson staining were performed on relevant tissues to analyze inflammation, and SOD activity was measured to assess oxidative stress levels. <b>Results:</b> The optimal drug-loaded hydrogel for chest drains contained 2% sodium alginate, 10% glycerol, and ropivacaine concentrations between 0.25% and 0.75%. This formulation showed superior morphological characteristics, degradation, and sustained-release properties. It also exhibited excellent bacteriostatic effects. The low-concentration (0.25%) drug-loaded hydrogel demonstrated better analgesic, anti-inflammatory, and oxidative stress-inhibitory effects in animal studies. <b>Conclusions:</b> The modified ropivacaine-alginate hydrogel-encapsulated chest drain offers a promising local slow-release strategy and may contribute to rapid rehabilitation in thoracic surgery.</p>","PeriodicalId":15138,"journal":{"name":"Journal of Biomaterials Applications","volume":" ","pages":"699-714"},"PeriodicalIF":2.5,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144955253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01Epub Date: 2025-09-04DOI: 10.1177/08853282251375172
Minoo Alavi, Mohammad Tabatabaei, Mohammad Tafazzoli-Shadpour, Mohamad Sadegh Aghajanzadeh
Mechanotransduction plays a pivotal role in shaping cellular behavior including migration, differentiation, and proliferation. To investigate this mechanism more accurately further, this study came up with a novel elastomeric substrate with a stiffness gradient using a sugar-based replica molding technique combined with a two-layer PDMS system. The efficient water solubility of candy allows easy release, creating a smooth substrate. By adjusting the substrate's thickness, the optimal effective gradient length for the study is achievable. Additionally, adjusting substrate thickness precisely controls stiffness, from very soft to hard-tissue-like rigidity. Atomic force microscopy characterization confirmed a continuous stiffness gradient on three commonly used PDMS mixtures, 1:30, 1:50, and 1:75, demonstrating the versatility of this method for fabricating and tuning substrates to mimic various tissue environments. In cellular experiments, 3T3 fibroblast cells exhibited a significant migratory response toward the 1:50/1:75 two-layer stiffness gradient, with cells migrating preferably in stiffer directions. Its cost-effectiveness, smooth surface, and ability to regulate gradient substrates with varied stiffness via different PDMS combinations are key advantages. By precisely replicating physiologically relevant mechanical microenvironments, this method advances mechanobiology research and facilitates modeling of stiffness-guided cellular behaviors, paving the way for reliable tissue engineering and regenerative medicine studies.
{"title":"Fabrication of a PDMS-based substrate with a stiffness gradient for modeling the mechanical microenvironment in single and collective cell studies.","authors":"Minoo Alavi, Mohammad Tabatabaei, Mohammad Tafazzoli-Shadpour, Mohamad Sadegh Aghajanzadeh","doi":"10.1177/08853282251375172","DOIUrl":"10.1177/08853282251375172","url":null,"abstract":"<p><p>Mechanotransduction plays a pivotal role in shaping cellular behavior including migration, differentiation, and proliferation. To investigate this mechanism more accurately further, this study came up with a novel elastomeric substrate with a stiffness gradient using a sugar-based replica molding technique combined with a two-layer PDMS system. The efficient water solubility of candy allows easy release, creating a smooth substrate. By adjusting the substrate's thickness, the optimal effective gradient length for the study is achievable. Additionally, adjusting substrate thickness precisely controls stiffness, from very soft to hard-tissue-like rigidity. Atomic force microscopy characterization confirmed a continuous stiffness gradient on three commonly used PDMS mixtures, 1:30, 1:50, and 1:75, demonstrating the versatility of this method for fabricating and tuning substrates to mimic various tissue environments. In cellular experiments, 3T3 fibroblast cells exhibited a significant migratory response toward the 1:50/1:75 two-layer stiffness gradient, with cells migrating preferably in stiffer directions. Its cost-effectiveness, smooth surface, and ability to regulate gradient substrates with varied stiffness via different PDMS combinations are key advantages. By precisely replicating physiologically relevant mechanical microenvironments, this method advances mechanobiology research and facilitates modeling of stiffness-guided cellular behaviors, paving the way for reliable tissue engineering and regenerative medicine studies.</p>","PeriodicalId":15138,"journal":{"name":"Journal of Biomaterials Applications","volume":" ","pages":"715-726"},"PeriodicalIF":2.5,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144992496","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01Epub Date: 2025-12-08DOI: 10.1007/s13205-025-04655-1
Preeti Joshi, Saumya, Faqua Zarreen, Haris Chandra Naik Gugulothu, M Moshahid Alam Rizvi, Bhavani Prasad Naik Nenavathu
Ag-Ca@CuO nanocomposites are fabricated using chemical precipitation method and are used for removing Cr (VI) and Pb (II) from the Yamuna River water in Delhi. Scanning electron microscopy confirmed the formation of rice grain-shaped CuO nanoparticles. Ag-Ca@CuO nanocomposites (CCA NCs) exhibited a surface area of 34.62 m²/g, notably superior to pristine CuO. At a concentration of 0.4 mg/mL of Ag-Ca@CuO nanocomposites, the highest removal rate of Lead (Pb) was observed to be 99.36%. For hexavalent chromium (Cr (VI)), the maximum removal efficiency was 72% under the same treatment conditions. Meanwhile, 63% of Nickel (Ni) removal is observed at 0.4 mg/mL treatment concentration. The incorporation of Ag and Ca played a crucial role in enhancing pollutant adsorption, suppressing electron-hole pair recombination, and promoting reactive oxygen species (ROS) generation for the degradation of toxic metal ions. We also studied the cytotoxic effects of CC and CCA NCs against the human HCT-116 cell line in a dose-dependent manner. At the nanocomposite's maximum concentration, i.e., 100 µg/mL, the cell viability for CC 2, CC 4, CCA 2 and CCA 4 was observed to be 47.64%, 35.29%, 19.83% and 8.88% respectively. IC50 value was also observed to be least for CCA 4 (17.71 µg/mL) followed by CCA 2 (31.61 µg/mL), CC 4 (72.93 µg/mL) and CC 2 (94.33 µg/mL). Cytotoxicity studies on human embryonic kidney (HEK 293) cell lines demonstrated minimal toxicity of the synthesised nanocomposites. This material could be used in wastewater treatment and as Drug-free therapeutics in cancer treatment.
Supplementary information: The online version contains supplementary material available at 10.1007/s13205-025-04655-1.
{"title":"Dual-functional Ag-Ca@CuO nanocomposites for heavy metal remediation and cancer cell inhibition.","authors":"Preeti Joshi, Saumya, Faqua Zarreen, Haris Chandra Naik Gugulothu, M Moshahid Alam Rizvi, Bhavani Prasad Naik Nenavathu","doi":"10.1007/s13205-025-04655-1","DOIUrl":"https://doi.org/10.1007/s13205-025-04655-1","url":null,"abstract":"<p><p>Ag-Ca@CuO nanocomposites are fabricated using chemical precipitation method and are used for removing Cr (VI) and Pb (II) from the Yamuna River water in Delhi. Scanning electron microscopy confirmed the formation of rice grain-shaped CuO nanoparticles. Ag-Ca@CuO nanocomposites (CCA NCs) exhibited a surface area of 34.62 m²/g, notably superior to pristine CuO. At a concentration of 0.4 mg/mL of Ag-Ca@CuO nanocomposites, the highest removal rate of Lead (Pb) was observed to be 99.36%. For hexavalent chromium (Cr (VI)), the maximum removal efficiency was 72% under the same treatment conditions. Meanwhile, 63% of Nickel (Ni) removal is observed at 0.4 mg/mL treatment concentration. The incorporation of Ag and Ca played a crucial role in enhancing pollutant adsorption, suppressing electron-hole pair recombination, and promoting reactive oxygen species (ROS) generation for the degradation of toxic metal ions. We also studied the cytotoxic effects of CC and CCA NCs against the human HCT-116 cell line in a dose-dependent manner. At the nanocomposite's maximum concentration, i.e., 100 µg/mL, the cell viability for CC 2, CC 4, CCA 2 and CCA 4 was observed to be 47.64%, 35.29%, 19.83% and 8.88% respectively. IC50 value was also observed to be least for CCA 4 (17.71 µg/mL) followed by CCA 2 (31.61 µg/mL), CC 4 (72.93 µg/mL) and CC 2 (94.33 µg/mL). Cytotoxicity studies on human embryonic kidney (HEK 293) cell lines demonstrated minimal toxicity of the synthesised nanocomposites. This material could be used in wastewater treatment and as Drug-free therapeutics in cancer treatment.</p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s13205-025-04655-1.</p>","PeriodicalId":7067,"journal":{"name":"3 Biotech","volume":"16 1","pages":"20"},"PeriodicalIF":2.9,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12686288/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145720321","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01Epub Date: 2025-12-08DOI: 10.1007/s13205-025-04632-8
Chenrui Ye, Mingyu Zhao, Yixin Mao, Meixuan Wu, Rong Zhou, Jun Du
While vasodilator-stimulated phosphoprotein (VASP) is a key cytoskeletal regulatory protein linked to oral squamous cell carcinoma development, its role in other cancers remains unexplored. In this study, we employed the TCGA database, ESTIMATE algorithm, and TIMER to investigate the correlations of VASP with survival outcomes, clinical features, and immune cell infiltration. We also utilized GO, KEGG, and GSEA enrichment analyses to explore its potential functions and constructed a PPI network using STRING and Cytoscape. Our pan-cancer analysis revealed that VASP mRNA was upregulated in ten and downregulated in six tumor types compared to normal tissues. Of particular interest, aberrant VASP expression was significantly associated with the progression and poor prognosis of liver hepatocellular carcinoma (LIHC) and lung adenocarcinoma (LUAD). The level of VASP mRNA showed notable upregulation in LIHC, and its expression was positively correlated with the levels of T-cell exhaustion markers. Univariate and multivariate Cox regression analysis indicated that VASP could serve as an independent diagnostic biomarker for this cancer type. Functional enrichment analysis revealed that VASP participates in several tumor-related processes, such as extracellular matrix degradation and the chemokine signaling pathway. In addition, VASP protein levels were observed to be significantly elevated in LUAD tissues compared to normal controls. VASP knockdown markedly suppressed the migratory capacity of LUAD cells in vitro. In conclusion, the aberrant expression of VASP is associated with poor prognosis in LIHC and LUAD, and VASP could be used as a novel predictive biomarker for LIHC patients.
{"title":"Pan-cancer analysis of oncogenic role of vasodilator-stimulated phosphoprotein (VASP) and validation in liver hepatocellular carcinoma.","authors":"Chenrui Ye, Mingyu Zhao, Yixin Mao, Meixuan Wu, Rong Zhou, Jun Du","doi":"10.1007/s13205-025-04632-8","DOIUrl":"https://doi.org/10.1007/s13205-025-04632-8","url":null,"abstract":"<p><p>While vasodilator-stimulated phosphoprotein (VASP) is a key cytoskeletal regulatory protein linked to oral squamous cell carcinoma development, its role in other cancers remains unexplored. In this study, we employed the TCGA database, ESTIMATE algorithm, and TIMER to investigate the correlations of VASP with survival outcomes, clinical features, and immune cell infiltration. We also utilized GO, KEGG, and GSEA enrichment analyses to explore its potential functions and constructed a PPI network using STRING and Cytoscape. Our pan-cancer analysis revealed that VASP mRNA was upregulated in ten and downregulated in six tumor types compared to normal tissues. Of particular interest, aberrant VASP expression was significantly associated with the progression and poor prognosis of liver hepatocellular carcinoma (LIHC) and lung adenocarcinoma (LUAD). The level of VASP mRNA showed notable upregulation in LIHC, and its expression was positively correlated with the levels of T-cell exhaustion markers. Univariate and multivariate Cox regression analysis indicated that VASP could serve as an independent diagnostic biomarker for this cancer type. Functional enrichment analysis revealed that VASP participates in several tumor-related processes, such as extracellular matrix degradation and the chemokine signaling pathway. In addition, VASP protein levels were observed to be significantly elevated in LUAD tissues compared to normal controls. VASP knockdown markedly suppressed the migratory capacity of LUAD cells in vitro. In conclusion, the aberrant expression of VASP is associated with poor prognosis in LIHC and LUAD, and VASP could be used as a novel predictive biomarker for LIHC patients.</p>","PeriodicalId":7067,"journal":{"name":"3 Biotech","volume":"16 1","pages":"22"},"PeriodicalIF":2.9,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12686284/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145720473","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}