首页 > 最新文献

Bioactive Materials最新文献

英文 中文
miR-423-5p-enriched small extracellular vesicles drive periodontal regeneration via Sfrp2+ cell expansion 富含mir -423-5p的细胞外小泡通过srp2 +细胞扩增驱动牙周再生
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-29 DOI: 10.1016/j.bioactmat.2025.11.026
Liya Ma , Yelin Zhang , Zijun Song , Pinwen Wang , Lu Liang , Mingzhu Chen , Xianmin Liao , Songtao Yang , Jiangtian Hu , Kai Chen , Hefeng Yang
Small extracellular vesicles (sEVs) show therapeutic potential for periodontitis but functional components remain unclear, limiting clinical periodontal therapy application. Identifying key bioactive molecules and enhancing their functions via engineering strategies may overcome these limitations. By comparing the periodontal tropism of sEVs from different stem cells and conducting functional miRNA profiling, we identified miR-423–5p as a key component for periodontal ligament cell (PDLC) osteogenic differentiation by targeting PLCB1 (Phospholipase C Beta 1). We engineered miR-423-5p–enriched sEVs (sEVsmiR−423−5p) with miRNA loading levels up to 100,000-fold higher than those of native sEVs. Compared with unmodified sEVs, sEVsmiR−423−5p promoted the formation of Sfrp2+ osteogenic fibroblasts at periodontal defect sites, ultimately facilitating early osteogenesis and regeneration of a native-like cementum–PDL–alveolar bone complex. These findings establish miR-423–5p as a pivotal osteoinductive effector in sEVs and demonstrate that its targeted enrichment markedly amplifies the regenerative capacity of sEVs, laying the groundwork for personalized nanovesicle-based regenerative therapies.
小细胞外囊泡(sev)显示出治疗牙周炎的潜力,但功能成分尚不清楚,限制了临床牙周治疗的应用。识别关键的生物活性分子并通过工程策略增强其功能可以克服这些限制。通过比较来自不同干细胞的sev的牙周向性并进行功能性miRNA分析,我们发现miR-423-5p是通过靶向PLCB1(磷脂酶C β 1)实现牙周韧带细胞(PDLC)成骨分化的关键成分。我们设计了mir -423-5p富集的sev (sEVsmiR -423-5p),其miRNA负载水平比天然sev高100,000倍。与未修饰的sev相比,sEVsmiR - 423 - 5p促进了牙周缺损部位sfp2 +成骨成纤维细胞的形成,最终促进了原生样牙骨质- pdl -牙槽骨复合体的早期成骨和再生。这些发现证实了miR-423-5p是sev中的关键骨诱导效应物,并证明其靶向富集显著增强了sev的再生能力,为个性化的基于纳米囊泡的再生疗法奠定了基础。
{"title":"miR-423-5p-enriched small extracellular vesicles drive periodontal regeneration via Sfrp2+ cell expansion","authors":"Liya Ma ,&nbsp;Yelin Zhang ,&nbsp;Zijun Song ,&nbsp;Pinwen Wang ,&nbsp;Lu Liang ,&nbsp;Mingzhu Chen ,&nbsp;Xianmin Liao ,&nbsp;Songtao Yang ,&nbsp;Jiangtian Hu ,&nbsp;Kai Chen ,&nbsp;Hefeng Yang","doi":"10.1016/j.bioactmat.2025.11.026","DOIUrl":"10.1016/j.bioactmat.2025.11.026","url":null,"abstract":"<div><div>Small extracellular vesicles (sEVs) show therapeutic potential for periodontitis but functional components remain unclear, limiting clinical periodontal therapy application. Identifying key bioactive molecules and enhancing their functions via engineering strategies may overcome these limitations. By comparing the periodontal tropism of sEVs from different stem cells and conducting functional miRNA profiling, we identified miR-423–5p as a key component for periodontal ligament cell (PDLC) osteogenic differentiation by targeting <em>PLCB1</em> (Phospholipase C Beta 1). We engineered miR-423-5p–enriched sEVs (sEVs<sup>miR−423−5p</sup>) with miRNA loading levels up to 100,000-fold higher than those of native sEVs. Compared with unmodified sEVs, sEVs<sup>miR−423−5p</sup> promoted the formation of Sfrp2<sup>+</sup> osteogenic fibroblasts at periodontal defect sites, ultimately facilitating early osteogenesis and regeneration of a native-like cementum–PDL–alveolar bone complex. These findings establish miR-423–5p as a pivotal osteoinductive effector in sEVs and demonstrate that its targeted enrichment markedly amplifies the regenerative capacity of sEVs, laying the groundwork for personalized nanovesicle-based regenerative therapies.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"58 ","pages":"Pages 19-32"},"PeriodicalIF":18.0,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145623193","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}
引用次数: 0
Sonogenic malate depleting modulator for tumor metabolic reprogramming and antitumor immune activation 肿瘤代谢重编程和抗肿瘤免疫激活的声源苹果酸消耗调节剂
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-28 DOI: 10.1016/j.bioactmat.2025.10.028
Run Yang , Bowen Li , Yun Fu , Chenxu Shang , Guoqing Feng , Xuheng Chen , Huining He , Zhengmian Zhang , Yang Bai , Bin Zheng
The malate/aspartate shuttle is essential for maintaining mitochondrial membrane potential (MMP) and supporting tumor metabolism and survival. However, developing effective, controllable strategies to manipulate malate metabolism in vivo remains a challenge. Here, we report a sonogenically activated malate depletion modulator (MDM), GO/BCT:Mn, which integrates graphene oxide (GO) with Ca/Mn co-doped barium titanate (BCT:Mn) nanoparticles, enabling simultaneous metabolic and immune modulation under ultrasonic stimulation. Mechanistic studies reveal ultrasound triggers spatial charge separation in GO/BCT:Mn, generating reductive electrons and oxidative holes. Electrons drive the reduction of H+ to H2, lowering MMP and providing a gas therapy effect, whereas oxidative holes convert NADH to NAD+, suppressing malate synthesis and disrupting the malate/aspartate shuttle, thereby impairing mitochondrial integrity. These synergistic actions induce mitochondrial depolarization, autophagy, and apoptosis. In a murine colon cancer model, treatment with GO/BCT:Mn markedly suppressed tumor cell proliferation (Ki67) and angiogenesis (VEGF, CD31), while promoting apoptosis (TUNEL, Caspase-3). Transcriptomic and flow cytometry analyses further revealed activation of immune-related pathways, accompanied by increased infiltration of CD4+/CD8+ T cells and mature dendritic cells, indicating that metabolic perturbation synergistically enhances anti-tumor immunity. Collectively, this work establishes a precise ultrasound-responsive nanoplatform that couples redox-mediated metabolic disruption with immune activation, offering a promising strategy for integrated metabolism–immune cancer therapy.
苹果酸/天冬氨酸穿梭对于维持线粒体膜电位(MMP)和支持肿瘤代谢和生存至关重要。然而,开发有效的、可控的策略来操纵苹果酸盐在体内的代谢仍然是一个挑战。在这里,我们报道了一种声源激活的苹果酸盐耗尽调节剂(MDM), GO/BCT:Mn,它将氧化石墨烯(GO)与Ca/Mn共掺杂的钛酸钡(BCT:Mn)纳米颗粒集成在一起,在超声波刺激下实现同时代谢和免疫调节。机理研究表明,超声触发GO/BCT:Mn中的空间电荷分离,产生还原电子和氧化空穴。电子驱动H+还原为H2,降低MMP并提供气体治疗效果,而氧化空穴将NADH转化为NAD+,抑制苹果酸合成并破坏苹果酸/天冬氨酸穿梭,从而损害线粒体完整性。这些协同作用诱导线粒体去极化、自噬和凋亡。在小鼠结肠癌模型中,氧化石墨烯/BCT:Mn治疗显著抑制肿瘤细胞增殖(Ki67)和血管生成(VEGF, CD31),同时促进细胞凋亡(TUNEL, Caspase-3)。转录组学和流式细胞术分析进一步揭示了免疫相关途径的激活,伴随着CD4+/CD8+ T细胞和成熟树突状细胞浸润的增加,表明代谢扰动协同增强抗肿瘤免疫。总的来说,这项工作建立了一个精确的超声响应纳米平台,将氧化还原介导的代谢破坏与免疫激活结合起来,为代谢-免疫癌症综合治疗提供了一个有希望的策略。
{"title":"Sonogenic malate depleting modulator for tumor metabolic reprogramming and antitumor immune activation","authors":"Run Yang ,&nbsp;Bowen Li ,&nbsp;Yun Fu ,&nbsp;Chenxu Shang ,&nbsp;Guoqing Feng ,&nbsp;Xuheng Chen ,&nbsp;Huining He ,&nbsp;Zhengmian Zhang ,&nbsp;Yang Bai ,&nbsp;Bin Zheng","doi":"10.1016/j.bioactmat.2025.10.028","DOIUrl":"10.1016/j.bioactmat.2025.10.028","url":null,"abstract":"<div><div>The malate/aspartate shuttle is essential for maintaining mitochondrial membrane potential (MMP) and supporting tumor metabolism and survival. However, developing effective, controllable strategies to manipulate malate metabolism in vivo remains a challenge. Here, we report a sonogenically activated malate depletion modulator (MDM), GO/BCT:Mn, which integrates graphene oxide (GO) with Ca/Mn co-doped barium titanate (BCT:Mn) nanoparticles, enabling simultaneous metabolic and immune modulation under ultrasonic stimulation. Mechanistic studies reveal ultrasound triggers spatial charge separation in GO/BCT:Mn, generating reductive electrons and oxidative holes. Electrons drive the reduction of H<sup>+</sup> to H<sub>2</sub>, lowering MMP and providing a gas therapy effect, whereas oxidative holes convert NADH to NAD<sup>+</sup>, suppressing malate synthesis and disrupting the malate/aspartate shuttle, thereby impairing mitochondrial integrity. These synergistic actions induce mitochondrial depolarization, autophagy, and apoptosis. In a murine colon cancer model, treatment with GO/BCT:Mn markedly suppressed tumor cell proliferation (Ki67) and angiogenesis (VEGF, CD31), while promoting apoptosis (TUNEL, Caspase-3). Transcriptomic and flow cytometry analyses further revealed activation of immune-related pathways, accompanied by increased infiltration of CD4<sup>+</sup>/CD8<sup>+</sup> T cells and mature dendritic cells, indicating that metabolic perturbation synergistically enhances anti-tumor immunity. Collectively, this work establishes a precise ultrasound-responsive nanoplatform that couples redox-mediated metabolic disruption with immune activation, offering a promising strategy for integrated metabolism–immune cancer therapy.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"56 ","pages":"Pages 682-702"},"PeriodicalIF":18.0,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145620755","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}
引用次数: 0
Hyaluronan regulates the assembly structure and biofunction of polycationic silk fibroin to boost microfracture 透明质酸调节聚阳离子丝素蛋白的组装结构和生物功能,促进微断裂
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-28 DOI: 10.1016/j.bioactmat.2025.11.037
Gong Li , Birui Yang , Zhulian Li , Peiyang Gu , Yaping Zou , Yuxiang Wang , Yujiang Fan , Yong Sun
Dysregulated inflammatory microenvironment and insufficient recruitment of functional cells/factors at the early stage limit the efficacy of microfracture (MF) therapy. Here, we regulated the assembly structure of functional polycationic silk fibroin (CSF) with sulfhydryl-modified hyaluronan (HS) to fabricate a cartilage repair patch (ECSF-HS-H), which achieved rapid hemostasis, recruitment of reparative cells/factors, and inflammation regulation. Through non-covalent/covalent interactions and β-sheet transitions, the assembly structure of CSF was reconfigured to adapt to the cyclic mechanical properties required under articular stress conditions while preserving inherent rapid hemostatic ability, reduce the potential inhibitory effect of cationic charges on cell proliferation, and enhance chondrogenic differentiation of stem cells. In a rabbit MF model, ECSF-HS-H demonstrated accelerated coagulation, reduced early inflammation in synovial fluid, promoted M2 macrophage polarization, and improved hyaline cartilage regeneration. This strategy of functional structural regulation at the molecular level offers a novel approach for developing protein-based scaffolds to enhance MF procedures.
炎症微环境失调和早期功能细胞/因子募集不足限制了微骨折(MF)治疗的疗效。本研究通过巯基修饰透明质酸(HS)调节功能性多阳离子丝素蛋白(CSF)的组装结构,制备软骨修复贴片(ECSF-HS-H),实现了快速止血、修复细胞/因子募集和炎症调节。通过非共价/共价相互作用和β-sheet转移,CSF的组装结构被重新配置,以适应关节应力条件下所需的循环力学性能,同时保持固有的快速止血能力,降低阳离子电荷对细胞增殖的潜在抑制作用,增强干细胞的软骨分化。在家兔MF模型中,ECSF-HS-H表现出加速凝血,减少滑膜液早期炎症,促进M2巨噬细胞极化,改善透明软骨再生。这种在分子水平上的功能结构调节策略为开发基于蛋白质的支架来增强MF程序提供了一种新的方法。
{"title":"Hyaluronan regulates the assembly structure and biofunction of polycationic silk fibroin to boost microfracture","authors":"Gong Li ,&nbsp;Birui Yang ,&nbsp;Zhulian Li ,&nbsp;Peiyang Gu ,&nbsp;Yaping Zou ,&nbsp;Yuxiang Wang ,&nbsp;Yujiang Fan ,&nbsp;Yong Sun","doi":"10.1016/j.bioactmat.2025.11.037","DOIUrl":"10.1016/j.bioactmat.2025.11.037","url":null,"abstract":"<div><div>Dysregulated inflammatory microenvironment and insufficient recruitment of functional cells/factors at the early stage limit the efficacy of microfracture (MF) therapy. Here, we regulated the assembly structure of functional polycationic silk fibroin (CSF) with sulfhydryl-modified hyaluronan (HS) to fabricate a cartilage repair patch (ECSF-HS-H), which achieved rapid hemostasis, recruitment of reparative cells/factors, and inflammation regulation. Through non-covalent/covalent interactions and β-sheet transitions, the assembly structure of CSF was reconfigured to adapt to the cyclic mechanical properties required under articular stress conditions while preserving inherent rapid hemostatic ability, reduce the potential inhibitory effect of cationic charges on cell proliferation, and enhance chondrogenic differentiation of stem cells. In a rabbit MF model, ECSF-HS-H demonstrated accelerated coagulation, reduced early inflammation in synovial fluid, promoted M2 macrophage polarization, and improved hyaline cartilage regeneration. This strategy of functional structural regulation at the molecular level offers a novel approach for developing protein-based scaffolds to enhance MF procedures.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"57 ","pages":"Pages 754-767"},"PeriodicalIF":18.0,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145621135","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}
引用次数: 0
Turmeric extracellular vesicles-derived “all-in-one” nanoagent enables full-cycle synergistic immunomodulation of lung cancer 姜黄细胞外囊泡衍生的“all-in-one”纳米剂可实现肺癌的全周期协同免疫调节
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-27 DOI: 10.1016/j.bioactmat.2025.10.017
Jichun Yang , Qianqian Wu , Yunqian Fu , Xiaohui Chen , Hengyi Chen , Yuhan Wang , Xiang Wu , Xin Cui , Sitong Wang , Yao Luo , Yufang Zhang , Yican Li , Yan Zhao , Zhixin Cha , Linke Shi , Xianlong Jiao , Fang Li , Yang Luo
Lung cancer's metastatic propensity and recurrence prevalence necessitate innovative immunotherapy strategies beyond conventional single-mode regulation. We engineered low-capacity turmeric-derived extracellular vesicles (TEVs) that integrated with zeolitic imidazolate framework-8 (ZIF-8) to construct an “all-in-one” nanoagent, addressing both high drug loading nanocarriers production and immunologically cold tumor challenges. The system co-delivered chlorin e6 (Ce6) and PD-L1 siRNA, while exploited TEVs' inherent curcumin for Wnt/β-catenin pathway inhibition. Ce6-mediated photodynamic therapy (PDT) induced immunogenic cell death (ICD), releasing damage associated molecular patterns (DAMPs) to activate antigen-presenting cells (APCs). Compared with control groups, artificial intelligence model confirmed the role of curcumin in enhancing immune infiltration by 6.1-fold. PD-L1 siRNA synergistically downregulates the checkpoint expression with a 66 % reduction in vivo to prevent the immune escape. This coordinated strategy achieved full-cycle immunomodulation: (1) ICD initiated antigens release, (2) Wnt/β-catenin pathway inhibition drived T cell infiltration, and (3) PD-L1 blockade receded the immune escape. In vivo results demonstrated that 64 % primary tumor suppression and 81 % metastasis reduction versus monotherapy groups. The ZIF-8@TEV hybrid platform exhibited 12.8 % payload loading efficiency, surpassing liposomal carriers by 4.7-fold. This study established a scalable nanoengineering approach to transform immunosuppressive tumors into immunotherapy-responsive targets through a full-cycle immune coordination.
肺癌的转移倾向和复发率需要创新的免疫治疗策略,而不是传统的单模调节。我们设计了低容量姜黄衍生的细胞外囊泡(TEVs),将其与沸石咪唑酸框架-8 (ZIF-8)结合,构建了一种“一体化”纳米剂,解决了高药物负载纳米载体的生产和免疫冷肿瘤挑战。该系统共递送氯e6 (Ce6)和PD-L1 siRNA,同时利用tev固有的姜黄素抑制Wnt/β-catenin途径。ce6介导的光动力疗法(PDT)诱导免疫原性细胞死亡(ICD),释放损伤相关分子模式(DAMPs)来激活抗原提呈细胞(APCs)。与对照组相比,人工智能模型证实姜黄素增强免疫浸润的作用提高了6.1倍。PD-L1 siRNA协同下调检查点表达,在体内减少66%,以防止免疫逃逸。这种协调策略实现了全周期免疫调节:(1)ICD启动抗原释放,(2)Wnt/β-catenin通路抑制驱动T细胞浸润,(3)PD-L1阻断减缓免疫逃逸。体内实验结果表明,与单药治疗组相比,原发肿瘤抑制率为64%,转移灶减少率为81%。ZIF-8@TEV混合平台的有效载荷效率为12.8%,比脂质体载体高出4.7倍。本研究建立了一种可扩展的纳米工程方法,通过全周期免疫协调将免疫抑制性肿瘤转化为免疫治疗应答性靶点。
{"title":"Turmeric extracellular vesicles-derived “all-in-one” nanoagent enables full-cycle synergistic immunomodulation of lung cancer","authors":"Jichun Yang ,&nbsp;Qianqian Wu ,&nbsp;Yunqian Fu ,&nbsp;Xiaohui Chen ,&nbsp;Hengyi Chen ,&nbsp;Yuhan Wang ,&nbsp;Xiang Wu ,&nbsp;Xin Cui ,&nbsp;Sitong Wang ,&nbsp;Yao Luo ,&nbsp;Yufang Zhang ,&nbsp;Yican Li ,&nbsp;Yan Zhao ,&nbsp;Zhixin Cha ,&nbsp;Linke Shi ,&nbsp;Xianlong Jiao ,&nbsp;Fang Li ,&nbsp;Yang Luo","doi":"10.1016/j.bioactmat.2025.10.017","DOIUrl":"10.1016/j.bioactmat.2025.10.017","url":null,"abstract":"<div><div>Lung cancer's metastatic propensity and recurrence prevalence necessitate innovative immunotherapy strategies beyond conventional single-mode regulation. We engineered low-capacity turmeric-derived extracellular vesicles (TEVs) that integrated with zeolitic imidazolate framework-8 (ZIF-8) to construct an “all-in-one” nanoagent, addressing both high drug loading nanocarriers production and immunologically cold tumor challenges. The system co-delivered chlorin e6 (Ce6) and PD-L1 siRNA, while exploited TEVs' inherent curcumin for Wnt/β-catenin pathway inhibition. Ce6-mediated photodynamic therapy (PDT) induced immunogenic cell death (ICD), releasing damage associated molecular patterns (DAMPs) to activate antigen-presenting cells (APCs). Compared with control groups, artificial intelligence model confirmed the role of curcumin in enhancing immune infiltration by 6.1-fold. PD-L1 siRNA synergistically downregulates the checkpoint expression with a 66 % reduction <em>in vivo</em> to prevent the immune escape. This coordinated strategy achieved full-cycle immunomodulation: (1) ICD initiated antigens release, (2) Wnt/β-catenin pathway inhibition drived T cell infiltration, and (3) PD-L1 blockade receded the immune escape. <em>In vivo</em> results demonstrated that 64 % primary tumor suppression and 81 % metastasis reduction versus monotherapy groups. The ZIF-8@TEV hybrid platform exhibited 12.8 % payload loading efficiency, surpassing liposomal carriers by 4.7-fold. This study established a scalable nanoengineering approach to transform immunosuppressive tumors into immunotherapy-responsive targets through a full-cycle immune coordination.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"56 ","pages":"Pages 666-681"},"PeriodicalIF":18.0,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145620756","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}
引用次数: 0
Three-tier framework for high-throughput biofabrication: Integrating 3D bioprinting, assistive platforms, and translational opportunities 高通量生物制造的三层框架:集成3D生物打印,辅助平台和转化机会
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-27 DOI: 10.1016/j.bioactmat.2025.11.024
Yogendra Pratap Singh , Joseph Christakiran Moses , Myoung Hwan Kim , Deepak Gupta , Vaibhav Pal , Irem Derman Deniz , Ethan Michael Gerhard , Ibrahim T. Ozbolat
The field of biofabrication is rapidly evolving, yet it faces persistent challenges, including long manufacturing latency, slow throughput, issues with reproducibility, and scalability limitations. High-throughput biofabrication (HTBF) has emerged as a powerful strategy which is presented here to address these gaps through a structured, three-tier framework. Tier 1 encompasses core HTBF methods, such as multi-modal bioprinting and robotic bioassembly, which enable the rapid fabrication of large, physiologically relevant tissue constructs. Tier 2 comprises assisting platforms, including microfluidics and microphysiological bioreactors, which provide perfusion, mechanical conditioning, multiplexable sensing, and process parallelization. Tier 3 represents HTBF outcomes, including organoids, organ-on-a-chip systems, and engineered tissue grafts that deliver clinically and pharmacologically relevant insights. These advancements enable the development of in-vitro models that streamline drug testing, making it more cost-effective and efficient, while enhancing the accuracy and reliability of preclinical drug evaluation. This review defines HTBF by outlining its core characteristics and framework, presenting insights into recent technological advancements and their applications in regenerative medicine and drug discovery. Additionally, it addresses the regulatory and clinical translation challenges that must be resolved to facilitate the adoption of HTBF in personalized healthcare.
生物制造领域正在迅速发展,但它面临着持续的挑战,包括制造延迟长、吞吐量低、可再现性问题和可扩展性限制。高通量生物制造(HTBF)已成为一种强大的战略,本文通过结构化的三层框架来解决这些差距。第1层包括核心HTBF方法,如多模态生物打印和机器人生物组装,可以快速制造大型生理相关组织结构。第2层包括辅助平台,包括微流体和微生理生物反应器,提供灌注、机械调节、可复用传感和过程并行化。Tier 3代表HTBF的结果,包括类器官、器官芯片系统和工程组织移植物,提供临床和药理学相关的见解。这些进步使体外模型的发展能够简化药物测试,使其更具成本效益和效率,同时提高临床前药物评估的准确性和可靠性。本文概述了HTBF的核心特征和框架,介绍了最近的技术进展及其在再生医学和药物发现中的应用。此外,它还解决了必须解决的监管和临床翻译挑战,以促进HTBF在个性化医疗保健中的采用。
{"title":"Three-tier framework for high-throughput biofabrication: Integrating 3D bioprinting, assistive platforms, and translational opportunities","authors":"Yogendra Pratap Singh ,&nbsp;Joseph Christakiran Moses ,&nbsp;Myoung Hwan Kim ,&nbsp;Deepak Gupta ,&nbsp;Vaibhav Pal ,&nbsp;Irem Derman Deniz ,&nbsp;Ethan Michael Gerhard ,&nbsp;Ibrahim T. Ozbolat","doi":"10.1016/j.bioactmat.2025.11.024","DOIUrl":"10.1016/j.bioactmat.2025.11.024","url":null,"abstract":"<div><div>The field of biofabrication is rapidly evolving, yet it faces persistent challenges, including long manufacturing latency, slow throughput, issues with reproducibility, and scalability limitations. High-throughput biofabrication (HTBF) has emerged as a powerful strategy which is presented here to address these gaps through a structured, three-tier framework. Tier 1 encompasses core HTBF methods, such as multi-modal bioprinting and robotic bioassembly, which enable the rapid fabrication of large, physiologically relevant tissue constructs. Tier 2 comprises assisting platforms, including microfluidics and microphysiological bioreactors, which provide perfusion, mechanical conditioning, multiplexable sensing, and process parallelization. Tier 3 represents HTBF outcomes, including organoids, organ-on-a-chip systems, and engineered tissue grafts that deliver clinically and pharmacologically relevant insights. These advancements enable the development of in-vitro models that streamline drug testing, making it more cost-effective and efficient, while enhancing the accuracy and reliability of preclinical drug evaluation. This review defines HTBF by outlining its core characteristics and framework, presenting insights into recent technological advancements and their applications in regenerative medicine and drug discovery. Additionally, it addresses the regulatory and clinical translation challenges that must be resolved to facilitate the adoption of HTBF in personalized healthcare.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"57 ","pages":"Pages 726-753"},"PeriodicalIF":18.0,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145621134","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}
引用次数: 0
A multifunctional hydrogel for obesity-associated tumor immunotherapy and postsurgical wound healing promotion 一种用于肥胖相关肿瘤免疫治疗和术后伤口愈合的多功能水凝胶
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-26 DOI: 10.1016/j.bioactmat.2025.11.027
Hongjuan Zhao , Di Meng , Yajing Wang , Yinke Wang , Qing Li , Yuxin Guo , Qingling Song , Lei Wang
The multidimensional complexity between metabolism and inflammation within the obese tumor microenvironment (OTME) poses substantial barriers to postsurgical immunotherapy and wound management. Herein, we engineered a multifunctional hydrogel (Lipo/CXB@Hydrogel) through covalent conjugation of dopamine-crosslinked oxidized hyaluronic acid and a ROS-sensitive linker, co-delivering the non-steroidal anti-inflammatory drug celecoxib (CXB) and the lipid metabolism modulator Lipofermata (Lipo) to facilitate T cell immunotherapy and wound healing. The unique multi-dynamic-bond crosslinked structure endows the hydrogel with excellent self-healing, tissue adhesiveness and mechanical properties. The implanted Lipo/CXB@Hydrogel degrades and releases CXB to suppress hyperinflammation and enhance intratumoral cytotoxic T lymphocyte (CTL) infiltration, while Lipo inhibits the predatory uptake of fatty acids by tumor cells in the OTME for competing metabolic resources of intratumoral infiltrated CTLs. Importantly, such a cascaded immunological effect of Lipo/CXB@Hydrogel treatment amplifies CTL proliferation and activity specifically through targeting the arachidonic acid (AA)/COX-2/PGE2 signaling axis, a central hub linking lipid metabolism and inflammation, initiating a long-lasting immune response to suppress colorectal tumor postsurgical recurrence and metastasis in obesity contexts. Moreover, the hydrogel can easily repeatedly close the reopened wounds and promote skin regeneration. Thus, this multifunctional hydrogel may provide a promising strategy for postsurgical obese tumor immunotherapy and wound closure.
肥胖肿瘤微环境(OTME)中代谢和炎症之间的多维复杂性给术后免疫治疗和伤口管理带来了实质性的障碍。在此,我们设计了一种多功能水凝胶(Lipo/CXB@Hydrogel),通过多巴胺交联氧化透明质酸和ros敏感连接物的共价偶联,共同递送非甾体抗炎药塞来昔布(CXB)和脂质代谢调节剂Lipofermata (Lipo),以促进T细胞免疫治疗和伤口愈合。独特的多动态键交联结构使水凝胶具有优异的自愈性、组织黏附性和力学性能。植入的Lipo/CXB@Hydrogel降解并释放CXB抑制高炎症并增强肿瘤内细胞毒性T淋巴细胞(CTL)浸润,同时Lipo抑制肿瘤细胞在OTME中掠夺性摄取脂肪酸,以争夺肿瘤内浸润的CTL的代谢资源。重要的是,Lipo/CXB@Hydrogel治疗的级联免疫效应通过靶向花生四烯酸(AA)/COX-2/PGE2信号轴(连接脂质代谢和炎症的中心枢纽)特异性地放大CTL增殖和活性,启动持久的免疫反应,抑制肥胖背景下结直肠肿瘤术后复发和转移。此外,水凝胶可以很容易地反复关闭重新打开的伤口,促进皮肤再生。因此,这种多功能水凝胶可能为肥胖肿瘤术后免疫治疗和伤口愈合提供一种有前景的策略。
{"title":"A multifunctional hydrogel for obesity-associated tumor immunotherapy and postsurgical wound healing promotion","authors":"Hongjuan Zhao ,&nbsp;Di Meng ,&nbsp;Yajing Wang ,&nbsp;Yinke Wang ,&nbsp;Qing Li ,&nbsp;Yuxin Guo ,&nbsp;Qingling Song ,&nbsp;Lei Wang","doi":"10.1016/j.bioactmat.2025.11.027","DOIUrl":"10.1016/j.bioactmat.2025.11.027","url":null,"abstract":"<div><div>The multidimensional complexity between metabolism and inflammation within the obese tumor microenvironment (OTME) poses substantial barriers to postsurgical immunotherapy and wound management. Herein, we engineered a multifunctional hydrogel (Lipo/CXB@Hydrogel) through covalent conjugation of dopamine-crosslinked oxidized hyaluronic acid and a ROS-sensitive linker, co-delivering the non-steroidal anti-inflammatory drug celecoxib (CXB) and the lipid metabolism modulator Lipofermata (Lipo) to facilitate T cell immunotherapy and wound healing. The unique multi-dynamic-bond crosslinked structure endows the hydrogel with excellent self-healing, tissue adhesiveness and mechanical properties. The implanted Lipo/CXB@Hydrogel degrades and releases CXB to suppress hyperinflammation and enhance intratumoral cytotoxic T lymphocyte (CTL) infiltration, while Lipo inhibits the predatory uptake of fatty acids by tumor cells in the OTME for competing metabolic resources of intratumoral infiltrated CTLs. Importantly, such a cascaded immunological effect of Lipo/CXB@Hydrogel treatment amplifies CTL proliferation and activity specifically through targeting the arachidonic acid (AA)/COX-2/PGE2 signaling axis, a central hub linking lipid metabolism and inflammation, initiating a long-lasting immune response to suppress colorectal tumor postsurgical recurrence and metastasis in obesity contexts. Moreover, the hydrogel can easily repeatedly close the reopened wounds and promote skin regeneration. Thus, this multifunctional hydrogel may provide a promising strategy for postsurgical obese tumor immunotherapy and wound closure.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"57 ","pages":"Pages 710-725"},"PeriodicalIF":18.0,"publicationDate":"2025-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145621133","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}
引用次数: 0
Magnesium ion implantation enhances the osseointegration and vascularization of 3D-Printed CoCrMo alloy scaffolds for load-bearing orthopedic applications 镁离子植入增强了3d打印CoCrMo合金支架的骨整合和血管化,用于承重骨科应用
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-26 DOI: 10.1016/j.bioactmat.2025.11.012
Ziyang Dong , Chenyuan Gao , Xinguang Wang , Ti Zhang , Xiao Geng , Jiazheng Chen , Junhao Feng , Yuhang Zheng , Yipu Zhang , Zhencan Han , Hao Wang , Ji Tan , Xianming Zhang , Yang Li , Zijian Li , Chongbin Wei , Qing Cai , Hua Tian
Total knee arthroplasty (TKA) remains the gold-standard treatment for end-stage osteoarthritis, yet persistent challenges in prosthetic material performance limit its long-term clinical efficacy. CoCrMo alloy is commonly used material of femoral component in TKA due to its excellent mechanical durability. However, two critical limitations persist: (1) substantial elastic modulus mismatch inducing stress-shielding effects, and (2) bioinert surface impairing osseointegration. To address these dual challenges, we developed a synergistic surface engineering strategy combining 3D-printed porous architecture with Mg2+ functionalization via plasma immersion ion implantation (PIII). The porous structure significantly reduced the elastic modulus and achieve biomimetic mechanical compatibility. Mg2+-implanted scaffolds (CoCrMo-Mg) demonstrated multifunctional bioactivity through synergistic physicochemical interactions. Surface topography modification via 3D printing generated micro-scale features that enhanced osteoblast adhesion through mechanotransduction pathways, while the release of Mg2+ exerted immunomodulatory, pro-angiogenic and osteogenic effects. Mg2+-mediated downregulation of pro-inflammatory cytokines, established an anti-inflammatory microenvironment conducive to bone regeneration, while Mg2+ stimulation promoted substantial neovascularization - collectively creating an osteogenic niche favoring coupled angiogenesis-osteogenesis process. These findings were further validated in vivo, where the CoCrMo-Mg scaffolds showed improved anti-inflammation, neovascularization and bone ingrowth capacities, along with favorable biomechanical integration. Overall, this dual-modality approach combining structural optimization with bioactive ion engineering establishes a paradigm for developing mechanically compliant and biologically active orthopedic implants, with particular translational relevance for cementless TKA applications.
全膝关节置换术(TKA)仍然是治疗终末期骨关节炎的金标准,但假体材料性能的持续挑战限制了其长期临床疗效。CoCrMo合金因其优异的机械耐久性成为TKA股骨假体常用材料。然而,两个关键的限制仍然存在:(1)大量的弹性模量不匹配导致应力屏蔽效应;(2)生物惰性表面损害骨整合。为了解决这些双重挑战,我们开发了一种协同表面工程策略,将3d打印多孔结构与通过等离子体浸泡离子注入(PIII)实现Mg2+功能化相结合。多孔结构显著降低弹性模量,实现仿生力学相容性。Mg2+植入支架(CoCrMo-Mg)通过协同的物理化学相互作用表现出多功能的生物活性。通过3D打印对表面形貌进行修饰,产生微尺度特征,通过机械转导途径增强成骨细胞粘附,同时释放Mg2+发挥免疫调节、促血管生成和成骨作用。Mg2+介导的促炎细胞因子的下调,建立了一个有利于骨再生的抗炎微环境,而Mg2+刺激促进了大量的新生血管——共同创造了一个有利于血管生成-成骨过程耦合的成骨生态位。这些发现在体内得到了进一步的验证,CoCrMo-Mg支架显示出更好的抗炎症、新生血管和骨长入能力,以及良好的生物力学整合。总的来说,这种将结构优化与生物活性离子工程相结合的双模方法为开发机械适应性和生物活性的骨科植入物建立了一个范例,特别是与无水泥TKA应用的转化相关。
{"title":"Magnesium ion implantation enhances the osseointegration and vascularization of 3D-Printed CoCrMo alloy scaffolds for load-bearing orthopedic applications","authors":"Ziyang Dong ,&nbsp;Chenyuan Gao ,&nbsp;Xinguang Wang ,&nbsp;Ti Zhang ,&nbsp;Xiao Geng ,&nbsp;Jiazheng Chen ,&nbsp;Junhao Feng ,&nbsp;Yuhang Zheng ,&nbsp;Yipu Zhang ,&nbsp;Zhencan Han ,&nbsp;Hao Wang ,&nbsp;Ji Tan ,&nbsp;Xianming Zhang ,&nbsp;Yang Li ,&nbsp;Zijian Li ,&nbsp;Chongbin Wei ,&nbsp;Qing Cai ,&nbsp;Hua Tian","doi":"10.1016/j.bioactmat.2025.11.012","DOIUrl":"10.1016/j.bioactmat.2025.11.012","url":null,"abstract":"<div><div>Total knee arthroplasty (TKA) remains the gold-standard treatment for end-stage osteoarthritis, yet persistent challenges in prosthetic material performance limit its long-term clinical efficacy. CoCrMo alloy is commonly used material of femoral component in TKA due to its excellent mechanical durability. However, two critical limitations persist: (1) substantial elastic modulus mismatch inducing stress-shielding effects, and (2) bioinert surface impairing osseointegration. To address these dual challenges, we developed a synergistic surface engineering strategy combining 3D-printed porous architecture with Mg<sup>2+</sup> functionalization <em>via</em> plasma immersion ion implantation (PIII). The porous structure significantly reduced the elastic modulus and achieve biomimetic mechanical compatibility. Mg<sup>2+</sup>-implanted scaffolds (CoCrMo-Mg) demonstrated multifunctional bioactivity through synergistic physicochemical interactions. Surface topography modification <em>via</em> 3D printing generated micro-scale features that enhanced osteoblast adhesion through mechanotransduction pathways, while the release of Mg<sup>2+</sup> exerted immunomodulatory, pro-angiogenic and osteogenic effects. Mg<sup>2+</sup>-mediated downregulation of pro-inflammatory cytokines, established an anti-inflammatory microenvironment conducive to bone regeneration, while Mg<sup>2+</sup> stimulation promoted substantial neovascularization - collectively creating an osteogenic niche favoring coupled angiogenesis-osteogenesis process. These findings were further validated <em>in vivo</em>, where the CoCrMo-Mg scaffolds showed improved anti-inflammation, neovascularization and bone ingrowth capacities, along with favorable biomechanical integration. Overall, this dual-modality approach combining structural optimization with bioactive ion engineering establishes a paradigm for developing mechanically compliant and biologically active orthopedic implants, with particular translational relevance for cementless TKA applications.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"57 ","pages":"Pages 676-693"},"PeriodicalIF":18.0,"publicationDate":"2025-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145621170","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}
引用次数: 0
Rapidly photocrosslinkable charged silk-based hydrogel for emergency hemostasis and multifunctional wound therapy 用于紧急止血和多功能伤口治疗的快速光交联带电丝基水凝胶
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-26 DOI: 10.1016/j.bioactmat.2025.11.022
Shiqi Wang , Jiaqi Wang , Mingyu Li , Dinghui Gao , Xin Yu , Penglong Zhao , Huan Dou , Chengchen Guo , Siming Yuan
Achieving rapid hemostasis, infection control, and tissue regeneration remains a central challenge in acute wound management, particularly under emergency and battlefield conditions. Current hemostatic materials often suffer from delayed coagulation, low bioactivity, and potential immunogenicity, limiting their use in critical care. Here, we present a chemically engineered charged silk-based hydrogel (SAMA) that integrates ultrafast gelation, intrinsic antibacterial activity, and programmable biological function. SAMA is synthesized via dual side-chain modification of silk fibroin with methacrylate (MA) for photo-crosslinking and dimethylamino groups (DMA) for surface charge tuning. The hydrogels enable photo-triggered crosslinking within 8 s under 405 nm light, while offering programmable surface charge and aqueous stability. The positively charged SAMA-5.0(+) hydrogel markedly enhances coagulation and platelet activation, as evidenced by reduced blood loss, shortened clotting time, and elevated CD62P+/PAC-1+ platelet populations. The hydrogels exhibit over 95 % antibacterial efficacy against Staphylococcus aureus and Escherichia coli, while maintaining excellent cytocompatibility, hemocompatibility, and promoting favorable responses in endothelial and immune cells. Mechanistically, SAMA(+) induces macrophage polarization toward the anti-inflammatory M2 phenotype, suppressing IL-6-mediated inflammation and facilitating angiogenesis and collagen remodeling. Controlled release assays demonstrate first-order kinetics for both the model protein (BSA) and ropivacaine, enabling sustained and timely delivery under neutral and mildly acidic wound environments. These findings position charged SAMA hydrogels as a clinically translatable solution for acute wound care, integrating rapid sealing, antimicrobial protection, immune regulation, and tissue repair into a low-cost platform suitable for wound and emergency applications.
实现快速止血、感染控制和组织再生仍然是急性伤口管理的核心挑战,特别是在紧急和战场条件下。目前的止血材料往往存在凝血延迟、生物活性低和潜在的免疫原性,限制了它们在重症监护中的应用。在这里,我们提出了一种化学工程带电丝基水凝胶(SAMA),它集成了超快速凝胶,固有的抗菌活性和可编程的生物功能。采用甲基丙烯酸酯(MA)进行光交联,二甲氨基(DMA)进行表面电荷调节,对丝素蛋白进行双侧链改性,合成了SAMA。该水凝胶在405 nm光下可在8秒内实现光触发交联,同时提供可编程的表面电荷和水稳定性。带正电荷的SAMA-5.0(+)水凝胶显著增强凝血和血小板活化,减少失血量,缩短凝血时间,提高CD62P+/PAC-1+血小板数量。该水凝胶对金黄色葡萄球菌和大肠杆菌的抗菌效果超过95%,同时保持良好的细胞相容性、血液相容性,促进内皮细胞和免疫细胞的良好反应。机制上,SAMA(+)诱导巨噬细胞向抗炎M2表型极化,抑制il -6介导的炎症,促进血管生成和胶原重塑。控制释放试验显示模型蛋白(BSA)和罗哌卡因的一级动力学,能够在中性和轻度酸性伤口环境下持续及时地递送。这些发现将充电SAMA水凝胶定位为急性伤口护理的临床可转化解决方案,将快速密封、抗菌保护、免疫调节和组织修复整合到一个适合伤口和紧急应用的低成本平台中。
{"title":"Rapidly photocrosslinkable charged silk-based hydrogel for emergency hemostasis and multifunctional wound therapy","authors":"Shiqi Wang ,&nbsp;Jiaqi Wang ,&nbsp;Mingyu Li ,&nbsp;Dinghui Gao ,&nbsp;Xin Yu ,&nbsp;Penglong Zhao ,&nbsp;Huan Dou ,&nbsp;Chengchen Guo ,&nbsp;Siming Yuan","doi":"10.1016/j.bioactmat.2025.11.022","DOIUrl":"10.1016/j.bioactmat.2025.11.022","url":null,"abstract":"<div><div>Achieving rapid hemostasis, infection control, and tissue regeneration remains a central challenge in acute wound management, particularly under emergency and battlefield conditions. Current hemostatic materials often suffer from delayed coagulation, low bioactivity, and potential immunogenicity, limiting their use in critical care. Here, we present a chemically engineered charged silk-based hydrogel (SAMA) that integrates ultrafast gelation, intrinsic antibacterial activity, and programmable biological function. SAMA is synthesized via dual side-chain modification of silk fibroin with methacrylate (MA) for photo-crosslinking and dimethylamino groups (DMA) for surface charge tuning. The hydrogels enable photo-triggered crosslinking within 8 s under 405 nm light, while offering programmable surface charge and aqueous stability. The positively charged SAMA-5.0(+) hydrogel markedly enhances coagulation and platelet activation, as evidenced by reduced blood loss, shortened clotting time, and elevated CD62P<sup>+</sup>/PAC-1<sup>+</sup> platelet populations. The hydrogels exhibit over 95 % antibacterial efficacy against <em>Staphylococcus aureus</em> and <em>Escherichia coli</em>, while maintaining excellent cytocompatibility, hemocompatibility, and promoting favorable responses in endothelial and immune cells. Mechanistically, SAMA(+) induces macrophage polarization toward the anti-inflammatory M2 phenotype, suppressing IL-6-mediated inflammation and facilitating angiogenesis and collagen remodeling. Controlled release assays demonstrate first-order kinetics for both the model protein (BSA) and ropivacaine, enabling sustained and timely delivery under neutral and mildly acidic wound environments. These findings position charged SAMA hydrogels as a clinically translatable solution for acute wound care, integrating rapid sealing, antimicrobial protection, immune regulation, and tissue repair into a low-cost platform suitable for wound and emergency applications.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"57 ","pages":"Pages 694-709"},"PeriodicalIF":18.0,"publicationDate":"2025-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145621169","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}
引用次数: 0
Robust adhesion between solid-state hydroxyapatite and bone tissue through surface demineralization 固体羟基磷灰石与骨组织之间通过表面脱矿的牢固粘附
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-25 DOI: 10.1016/j.bioactmat.2025.11.030
Shichao Xie , Masahiro Okada , Haruyuki Aoyagi , Akihisa Otaka , Xiaofeng Yang , Takayoshi Nakano , Takuya Matsumoto

Objective

Current bone adhesives typically lack adequate mechanical strength, long-term stability, or biocompatibility. To address these limitations, we designed a new adhesion strategy using a solid-state hydroxyapatite (HAp) adhesive in combination with bone surface demineralization.

Methods

Solid-state HAp adhesives were synthesized via wet chemical precipitation and heat treatment. Cortical bone specimens were partially demineralized with phosphoric acid (H3PO4) or ethylenediaminetetraacetic acid (EDTA), and characterized using scanning electron microscopy (SEM) and attenuated total reflectance–Fourier transform infrared spectroscopy (ATR-FTIR). Shear adhesion strength of HAp to demineralized bone was measured over time. In vivo fixation was assessed in rats using micro-computed tomography and histology. Statistical analysis used Tukey-Kramer tests after normality and variance checks.

Results

Although the HAp adhesive failed to adhere to non-demineralized bone, effective adhesion was achieved on the surface-demineralized bone tissue. Shear adhesion strength was significantly higher in EDTA-treated samples (238.4 kPa at 10 h) compared to H3PO4-treated samples (102.9 kPa at 1 h), with performance correlating with demineralization depth. ATR-FTIR and SEM analyses revealed that EDTA preserved collagen's triple-helix structure and free water content, both enhancing adhesion. Animal experiments confirmed stable fixation of HAp adhesive to demineralized bone tissue.

Conclusions

Surface demineralization enabled strong adhesion of the solid-state HAp adhesive to bone by exposing collagen swollen with water. Adhesion strength was influenced by structural changes in the demineralized layer, and the adhesive provided stable in vivo fixation, supporting its potential for bone-anchored biomedical applications.
目前的骨胶粘剂通常缺乏足够的机械强度、长期稳定性或生物相容性。为了解决这些限制,我们设计了一种新的粘连策略,使用固态羟基磷灰石(HAp)粘接剂结合骨表面脱矿。方法采用湿法化学沉淀法和热处理法制备固态HAp胶粘剂。用磷酸(H3PO4)或乙二胺四乙酸(EDTA)对皮质骨标本进行部分脱矿处理,并用扫描电镜(SEM)和衰减全反射-傅里叶变换红外光谱(ATR-FTIR)对其进行表征。随时间测定HAp与脱矿骨的剪切粘附强度。使用显微计算机断层扫描和组织学评估大鼠体内固定。经正态性和方差检验后采用Tukey-Kramer检验进行统计分析。结果羟基磷灰石胶粘剂在非脱矿骨上不能粘附,但在表面脱矿骨组织上能有效粘附。edta处理样品的剪切黏附强度(10h时为238.4 kPa)显著高于h3po4处理样品(1h时为102.9 kPa),其性能与脱矿深度相关。ATR-FTIR和SEM分析表明,EDTA保留了胶原蛋白的三螺旋结构和游离水含量,增强了粘连。动物实验证实HAp粘接剂对脱矿骨组织的固定稳定。结论表面脱矿使固态羟基磷灰石胶粘剂通过暴露被水肿胀的胶原蛋白使其与骨具有较强的粘附性。脱矿层的结构变化影响粘接剂的粘附强度,该粘接剂提供稳定的体内固定,支持其骨锚定生物医学应用的潜力。
{"title":"Robust adhesion between solid-state hydroxyapatite and bone tissue through surface demineralization","authors":"Shichao Xie ,&nbsp;Masahiro Okada ,&nbsp;Haruyuki Aoyagi ,&nbsp;Akihisa Otaka ,&nbsp;Xiaofeng Yang ,&nbsp;Takayoshi Nakano ,&nbsp;Takuya Matsumoto","doi":"10.1016/j.bioactmat.2025.11.030","DOIUrl":"10.1016/j.bioactmat.2025.11.030","url":null,"abstract":"<div><h3>Objective</h3><div>Current bone adhesives typically lack adequate mechanical strength, long-term stability, or biocompatibility. To address these limitations, we designed a new adhesion strategy using a solid-state hydroxyapatite (HAp) adhesive in combination with bone surface demineralization.</div></div><div><h3>Methods</h3><div>Solid-state HAp adhesives were synthesized via wet chemical precipitation and heat treatment. Cortical bone specimens were partially demineralized with phosphoric acid (H<sub>3</sub>PO<sub>4</sub>) or ethylenediaminetetraacetic acid (EDTA), and characterized using scanning electron microscopy (SEM) and attenuated total reflectance–Fourier transform infrared spectroscopy (ATR-FTIR). Shear adhesion strength of HAp to demineralized bone was measured over time. <em>In vivo</em> fixation was assessed in rats using micro-computed tomography and histology. Statistical analysis used Tukey-Kramer tests after normality and variance checks.</div></div><div><h3>Results</h3><div>Although the HAp adhesive failed to adhere to non-demineralized bone, effective adhesion was achieved on the surface-demineralized bone tissue. Shear adhesion strength was significantly higher in EDTA-treated samples (238.4 kPa at 10 h) compared to H<sub>3</sub>PO<sub>4</sub>-treated samples (102.9 kPa at 1 h), with performance correlating with demineralization depth. ATR-FTIR and SEM analyses revealed that EDTA preserved collagen's triple-helix structure and free water content, both enhancing adhesion. Animal experiments confirmed stable fixation of HAp adhesive to demineralized bone tissue.</div></div><div><h3>Conclusions</h3><div>Surface demineralization enabled strong adhesion of the solid-state HAp adhesive to bone by exposing collagen swollen with water. Adhesion strength was influenced by structural changes in the demineralized layer, and the adhesive provided stable <em>in vivo</em> fixation, supporting its potential for bone-anchored biomedical applications.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"57 ","pages":"Pages 632-645"},"PeriodicalIF":18.0,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145621131","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}
引用次数: 0
Modular hydrogel niche orchestrates microenvironment detoxification and stem cell redirecting toward precision osteoarthritis therapy 模块化水凝胶生态位协调微环境解毒和干细胞重定向,以精确治疗骨关节炎
IF 18 1区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2025-11-25 DOI: 10.1016/j.bioactmat.2025.11.023
Yang Huang , Yan Wang , Guoyu Yang , Yunsheng Jiang , Menglong Sun , Shidi Cheng , Ling Zhong , Xiangling Pu , Bin Yang , Jiajia Chen , Zhiyu Chen , Chenwen Li , Liu Yang , Cheng Chen , Jianxiang Zhang
Osteoarthritis (OA), a debilitating degenerative joint disease driven by chronic inflammation and cartilage degradation, remains inadequately addressed by current therapies. While mesenchymal stem cells (MSCs) offer regenerative potential, their clinical efficacy is hindered by poor survival, transient retention, and pathological joint microenvironments. Here, we present a bioengineered hydrogel-based stem cell niche (PPT hydrogel) that dynamically coordinates microenvironmental reprogramming and MSC functional redirection to achieve sustained OA treatment. The PPT platform integrates intrinsic antioxidative and anti-inflammatory properties to neutralize OA-associated oxidative stress and inflammation while enhancing bone marrow MSCs (BMSCs) survival via Hedgehog signaling activation, overcoming limitations of conventional cell delivery. By incorporating a pro-differentiation agent, PPT hydrogel steers BMSCs toward stable hyaline cartilage regeneration, suppressing the formation of fibrotic and hypertrophic cartilage even under inflammatory conditions. Furthermore, PPT amplifies BMSCs paracrine signaling to restore redox homeostasis and autophagy flux in resident chondrocytes through FOXO1-dependent mechanisms, establishing a self-reinforcing therapeutic loop. The modular amphiphilic design enables spatiotemporal co-delivery of diverse therapeutics, synergistically regulating stem cell behavior and host tissue responses. Through bridging stem cell-chondrocyte crosstalk, this multifaceted PPT hydrogel represents a transformative paradigm for precision regenerative medicine, offering a universally adaptable platform to address complex inflammatory and degenerative diseases beyond OA.
骨关节炎(OA)是一种由慢性炎症和软骨退化引起的衰弱性退行性关节疾病,目前的治疗方法仍未充分解决。虽然间充质干细胞(MSCs)具有再生潜力,但其临床疗效受到生存不良、短暂滞留和病理关节微环境的阻碍。在这里,我们提出了一种生物工程水凝胶干细胞生态位(PPT水凝胶),它可以动态协调微环境重编程和MSC功能重定向,以实现持续的OA治疗。PPT平台整合了固有的抗氧化和抗炎特性,以中和oa相关的氧化应激和炎症,同时通过Hedgehog信号激活提高骨髓间充质干细胞(BMSCs)的存活率,克服了传统细胞递送的局限性。通过加入促分化剂,PPT水凝胶引导骨髓间充质干细胞走向稳定的透明软骨再生,即使在炎症条件下也能抑制纤维化和增生性软骨的形成。此外,PPT通过fox01依赖机制,放大骨髓间充质干细胞旁分泌信号,恢复常驻软骨细胞的氧化还原稳态和自噬通量,建立一个自我强化的治疗循环。模块化的两亲性设计使多种治疗方法的时空共递送,协同调节干细胞行为和宿主组织反应。通过桥接干细胞-软骨串音,这种多面体PPT水凝胶代表了精确再生医学的变革范例,为解决OA以外的复杂炎症和退行性疾病提供了一个普遍适应性的平台。
{"title":"Modular hydrogel niche orchestrates microenvironment detoxification and stem cell redirecting toward precision osteoarthritis therapy","authors":"Yang Huang ,&nbsp;Yan Wang ,&nbsp;Guoyu Yang ,&nbsp;Yunsheng Jiang ,&nbsp;Menglong Sun ,&nbsp;Shidi Cheng ,&nbsp;Ling Zhong ,&nbsp;Xiangling Pu ,&nbsp;Bin Yang ,&nbsp;Jiajia Chen ,&nbsp;Zhiyu Chen ,&nbsp;Chenwen Li ,&nbsp;Liu Yang ,&nbsp;Cheng Chen ,&nbsp;Jianxiang Zhang","doi":"10.1016/j.bioactmat.2025.11.023","DOIUrl":"10.1016/j.bioactmat.2025.11.023","url":null,"abstract":"<div><div>Osteoarthritis (OA), a debilitating degenerative joint disease driven by chronic inflammation and cartilage degradation, remains inadequately addressed by current therapies. While mesenchymal stem cells (MSCs) offer regenerative potential, their clinical efficacy is hindered by poor survival, transient retention, and pathological joint microenvironments. Here, we present a bioengineered hydrogel-based stem cell niche (PPT hydrogel) that dynamically coordinates microenvironmental reprogramming and MSC functional redirection to achieve sustained OA treatment. The PPT platform integrates intrinsic antioxidative and anti-inflammatory properties to neutralize OA-associated oxidative stress and inflammation while enhancing bone marrow MSCs (BMSCs) survival via Hedgehog signaling activation, overcoming limitations of conventional cell delivery. By incorporating a pro-differentiation agent, PPT hydrogel steers BMSCs toward stable hyaline cartilage regeneration, suppressing the formation of fibrotic and hypertrophic cartilage even under inflammatory conditions. Furthermore, PPT amplifies BMSCs paracrine signaling to restore redox homeostasis and autophagy flux in resident chondrocytes through FOXO1-dependent mechanisms, establishing a self-reinforcing therapeutic loop. The modular amphiphilic design enables spatiotemporal co-delivery of diverse therapeutics, synergistically regulating stem cell behavior and host tissue responses. Through bridging stem cell-chondrocyte crosstalk, this multifaceted PPT hydrogel represents a transformative paradigm for precision regenerative medicine, offering a universally adaptable platform to address complex inflammatory and degenerative diseases beyond OA.</div></div>","PeriodicalId":8762,"journal":{"name":"Bioactive Materials","volume":"57 ","pages":"Pages 660-675"},"PeriodicalIF":18.0,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145621130","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}
引用次数: 0
期刊
Bioactive Materials
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1