首页 > 最新文献

ACS Biomaterials Science & Engineering最新文献

英文 中文
Extracellular Vesicles Derived from Neutrophils Accelerate Bone Regeneration by Promoting Osteogenic Differentiation of BMSCs 中性粒细胞衍生的细胞外小泡通过促进 BMSCs 的成骨分化加速骨再生
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-05-04 DOI: 10.1021/acsbiomaterials.4c00106
Le Wang, Guanhua Zhang, Ye Gao, Taiqiang Dai, Jie Yu, Ya Liu, Han Bao, Jianzhen She, Yan Hou, Liang Kong* and Bolei Cai*, 

The reconstruction of bone defects has been associated with severe challenges worldwide. Nowadays, bone marrow mesenchymal stem cell (BMSC)-based cell sheets have rendered this approach a promising way to facilitate osteogenic regeneration in vivo. Extracellular vesicles (EVs) play an essential role in intercellular communication and execution of various biological functions and are often employed as an ideal natural endogenous nanomedicine for restoring the structure and functions of damaged tissues. The perception of polymorphonuclear leukocytes (neutrophils, PMNs) as indiscriminate killer cells is gradually changing, with new evidence suggesting a role for these cells in tissue repair and regeneration, particularly in the context of bone healing. However, the role of EVs derived from PMNs (PMN-EVs) in bone regeneration remains largely unknown, with limited research being conducted on this aspect. In the current study, we investigated the effects of PMN-EVs on BMSCs and the underlying molecular mechanisms as well as the potential application of PMN-EVs in bone regeneration. Toward this end, BMSC-based cell sheets with integrated PMN-EVs (BS@PMN-EVs) were developed for bone defect regeneration. PMN-EVs were found to significantly enhance the proliferation and osteogenic differentiation of BMSCs in vitro. Furthermore, BS@PMN-EVs were found to significantly accelerate bone regeneration in vivo by enhancing the maturation of the newly formed bone in rat calvarial defects; this is likely attributable to the effect of PMN-EVs in promoting the expression of key osteogenic proteins such as SOD2 and GJA1 in BMSCs. In conclusion, our findings demonstrate the crucial role of PMN-EVs in promoting the osteogenic differentiation of BMSCs during bone regeneration. Furthermore, this study proposes a novel strategy for enhancing bone repair and regeneration via the integration of PMN-EVs with BMSC-based cell sheets.

骨缺损的重建一直是全世界面临的严峻挑战。如今,以骨髓间充质干细胞(BMSC)为基础的细胞薄片已成为促进体内骨再生的一种有前途的方法。细胞外囊泡(EVs)在细胞间通信和执行各种生物功能方面发挥着重要作用,通常被用作恢复受损组织结构和功能的理想天然内源性纳米药物。人们对多形核白细胞(嗜中性粒细胞,PMNs)作为滥杀细胞的看法正在逐渐改变,新的证据表明这些细胞在组织修复和再生中发挥作用,尤其是在骨愈合方面。然而,从 PMNs 提取的 EVs(PMN-EVs)在骨再生中的作用在很大程度上仍不为人所知,这方面的研究也很有限。在本研究中,我们探讨了 PMN-EVs 对 BMSCs 的影响及其分子机制,以及 PMN-EVs 在骨再生中的潜在应用。为此,我们开发了整合了 PMN-EVs 的基于 BMSC 的细胞片(BS@PMN-EVs),用于骨缺损再生。研究发现,PMN-EVs 能显著增强 BMSCs 在体外的增殖和成骨分化。此外,研究还发现,BS@PMN-EVs 还能提高大鼠腓骨缺损处新形成骨的成熟度,从而明显加速体内骨再生;这可能是由于 PMN-EVs 能促进 BMSCs 中关键成骨蛋白(如 SOD2 和 GJA1)的表达。总之,我们的研究结果证明了 PMN-EV 在骨再生过程中促进 BMSCs 成骨分化的关键作用。此外,本研究还提出了一种新策略,即通过将 PMN-EV 与基于 BMSC 的细胞薄片相结合来增强骨修复和再生。
{"title":"Extracellular Vesicles Derived from Neutrophils Accelerate Bone Regeneration by Promoting Osteogenic Differentiation of BMSCs","authors":"Le Wang,&nbsp;Guanhua Zhang,&nbsp;Ye Gao,&nbsp;Taiqiang Dai,&nbsp;Jie Yu,&nbsp;Ya Liu,&nbsp;Han Bao,&nbsp;Jianzhen She,&nbsp;Yan Hou,&nbsp;Liang Kong* and Bolei Cai*,&nbsp;","doi":"10.1021/acsbiomaterials.4c00106","DOIUrl":"10.1021/acsbiomaterials.4c00106","url":null,"abstract":"<p >The reconstruction of bone defects has been associated with severe challenges worldwide. Nowadays, bone marrow mesenchymal stem cell (BMSC)-based cell sheets have rendered this approach a promising way to facilitate osteogenic regeneration <i>in vivo</i>. Extracellular vesicles (EVs) play an essential role in intercellular communication and execution of various biological functions and are often employed as an ideal natural endogenous nanomedicine for restoring the structure and functions of damaged tissues. The perception of polymorphonuclear leukocytes (neutrophils, PMNs) as indiscriminate killer cells is gradually changing, with new evidence suggesting a role for these cells in tissue repair and regeneration, particularly in the context of bone healing. However, the role of EVs derived from PMNs (PMN-EVs) in bone regeneration remains largely unknown, with limited research being conducted on this aspect. In the current study, we investigated the effects of PMN-EVs on BMSCs and the underlying molecular mechanisms as well as the potential application of PMN-EVs in bone regeneration. Toward this end, BMSC-based cell sheets with integrated PMN-EVs (BS@PMN-EVs) were developed for bone defect regeneration. PMN-EVs were found to significantly enhance the proliferation and osteogenic differentiation of BMSCs <i>in vitro</i>. Furthermore, BS@PMN-EVs were found to significantly accelerate bone regeneration <i>in vivo</i> by enhancing the maturation of the newly formed bone in rat calvarial defects; this is likely attributable to the effect of PMN-EVs in promoting the expression of key osteogenic proteins such as SOD2 and GJA1 in BMSCs. In conclusion, our findings demonstrate the crucial role of PMN-EVs in promoting the osteogenic differentiation of BMSCs during bone regeneration. Furthermore, this study proposes a novel strategy for enhancing bone repair and regeneration via the integration of PMN-EVs with BMSC-based cell sheets.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acsbiomaterials.4c00106","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140833617","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multifunctional Drug- and AuNRs-Loaded ROS-Responsive Selenium-Containing Polyurethane Nanofibers for Smart Wound Healing 用于智能伤口愈合的多功能药物和 AuNR 负载 ROS 响应型含硒聚氨酯纳米纤维
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-05-03 DOI: 10.1021/acsbiomaterials.4c00363
Wajiha Ahmed, Shifen Li, Min Liang, Yongyuan Kang, Xiaoqing Liu and Changyou Gao*, 

Elevated levels of ROS, bacterial infection, inflammation, and improper regeneration are the factors that need to be addressed simultaneously for achieving effective wound healing without scar formation. This study focuses on the fabrication of electrospun ROS-responsive selenium-containing polyurethane nanofibers incorporating deferoxamine mesylate (Def), indomethacin (Indo), and gold nanorods (AuNRs) as proangiogenesis, anti-inflammatory, and antibacterial agents for synchronized delivery to a full-thickness wound in vivo. The structure of the fabricated nanofibers was analyzed by various techniques. Toxicity was checked by CCK-8 and hemolytic assays. The efficiency of wound healing in vitro was verified by a transwell assay and cell scratch assay. The wound healing efficiency of the nanofibers was assayed in full-thickness wounds in a rat model. The multifunctional nanofibers had a porous structure, enhanced antioxidation, antibacterial activity, and promoted wound healing. They eradicated TNF-α and IL-6, increased IL-10 expression, and revealed the angiogenic potential by increased expression of HIF-1α, VEGF, and CD31.

ROS 水平升高、细菌感染、炎症和不当再生是实现有效伤口愈合而不形成疤痕需要同时解决的因素。本研究的重点是制造电纺 ROS 响应型含硒聚氨酯纳米纤维,将甲磺酸去氧胺(Def)、吲哚美辛(Indo)和金纳米棒(AuNRs)作为促进血管生成、抗炎和抗菌剂,同步输送到体内全厚伤口。通过各种技术分析了制备的纳米纤维的结构。毒性通过 CCK-8 和溶血试验进行检测。体外伤口愈合的效率通过透孔试验和细胞划痕试验进行了验证。纳米纤维的伤口愈合效率在大鼠模型的全厚伤口中进行了检测。多功能纳米纤维具有多孔结构,能增强抗氧化和抗菌活性,促进伤口愈合。它们消除了 TNF-α 和 IL-6,增加了 IL-10 的表达,并通过增加 HIF-1α、VEGF 和 CD31 的表达显示了血管生成潜力。
{"title":"Multifunctional Drug- and AuNRs-Loaded ROS-Responsive Selenium-Containing Polyurethane Nanofibers for Smart Wound Healing","authors":"Wajiha Ahmed,&nbsp;Shifen Li,&nbsp;Min Liang,&nbsp;Yongyuan Kang,&nbsp;Xiaoqing Liu and Changyou Gao*,&nbsp;","doi":"10.1021/acsbiomaterials.4c00363","DOIUrl":"10.1021/acsbiomaterials.4c00363","url":null,"abstract":"<p >Elevated levels of ROS, bacterial infection, inflammation, and improper regeneration are the factors that need to be addressed simultaneously for achieving effective wound healing without scar formation. This study focuses on the fabrication of electrospun ROS-responsive selenium-containing polyurethane nanofibers incorporating deferoxamine mesylate (Def), indomethacin (Indo), and gold nanorods (AuNRs) as proangiogenesis, anti-inflammatory, and antibacterial agents for synchronized delivery to a full-thickness wound <i>in vivo</i>. The structure of the fabricated nanofibers was analyzed by various techniques. Toxicity was checked by CCK-8 and hemolytic assays. The efficiency of wound healing <i>in vitro</i> was verified by a transwell assay and cell scratch assay. The wound healing efficiency of the nanofibers was assayed in full-thickness wounds in a rat model. The multifunctional nanofibers had a porous structure, enhanced antioxidation, antibacterial activity, and promoted wound healing. They eradicated TNF-α and IL-6, increased IL-10 expression, and revealed the angiogenic potential by increased expression of HIF-1α, VEGF, and CD31.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140842481","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Gold Nanoparticles Confined in Mesoporous Bioactive Glass for Periodontitis Therapy 用于牙周炎治疗的介孔生物活性玻璃中的纳米金粒子
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-05-03 DOI: 10.1021/acsbiomaterials.4c00107
Dong Zhou, Min Ge, QiHui Wang, Jingru Sun, Haiyan Yao, Yunyun Deng, Lan Xiao, Jiaolong Wang* and Junchao Wei*, 

Periodontitis is a chronic disease caused by bacterial infection and is characterized with alveolar bone resorption. Bone regeneration in periodontitis remains a critical challenge because bacterial infection induced an unfavorable microenvironment for osteogenesis. Therefore, it is necessary to design proper therapeutic platforms to control bacterial infection and promote bone regeneration. Herein, mesoporous bioactive glass (MBG) with different pore sizes (3.0, 4.3, and 12.3 nm) was used as an in situ reactor to confine the growth of gold nanoparticles (Au NPs), forming MBG@Au hybrids which combine the osteoconductivity of MBG and antibacterial properties of Au NPs. Upon near-infrared (NIR) irradiation, the MBG@Au NPs showed efficient antibacterial properties both in vitro and in vivo. Besides, the osteogenesis properties of MBG@Au also improved under NIR irradiation. Furthermore, the in vivo results demonstrated that MBG@Au can effectively promote alveolar bone regeneration and realize the healing of serious periodontitis.

牙周炎是一种由细菌感染引起的慢性疾病,以牙槽骨吸收为特征。由于细菌感染会诱发不利于骨生成的微环境,因此牙周炎的骨再生仍然是一项严峻的挑战。因此,有必要设计适当的治疗平台来控制细菌感染和促进骨再生。在此,研究人员使用不同孔径(3.0、4.3 和 12.3 nm)的介孔生物活性玻璃(MBG)作为原位反应器,限制金纳米粒子(Au NPs)的生长,形成 MBG@Au 混合物,该混合物结合了 MBG 的骨传导性和 Au NPs 的抗菌特性。在近红外(NIR)照射下,MBG@Au NPs 在体外和体内都表现出高效的抗菌特性。此外,在近红外照射下,MBG@Au 的成骨特性也得到了改善。此外,体内研究结果表明,MBG@Au 能有效促进牙槽骨再生,实现严重牙周炎的治愈。
{"title":"Gold Nanoparticles Confined in Mesoporous Bioactive Glass for Periodontitis Therapy","authors":"Dong Zhou,&nbsp;Min Ge,&nbsp;QiHui Wang,&nbsp;Jingru Sun,&nbsp;Haiyan Yao,&nbsp;Yunyun Deng,&nbsp;Lan Xiao,&nbsp;Jiaolong Wang* and Junchao Wei*,&nbsp;","doi":"10.1021/acsbiomaterials.4c00107","DOIUrl":"10.1021/acsbiomaterials.4c00107","url":null,"abstract":"<p >Periodontitis is a chronic disease caused by bacterial infection and is characterized with alveolar bone resorption. Bone regeneration in periodontitis remains a critical challenge because bacterial infection induced an unfavorable microenvironment for osteogenesis. Therefore, it is necessary to design proper therapeutic platforms to control bacterial infection and promote bone regeneration. Herein, mesoporous bioactive glass (MBG) with different pore sizes (3.0, 4.3, and 12.3 nm) was used as an in situ reactor to confine the growth of gold nanoparticles (Au NPs), forming MBG@Au hybrids which combine the osteoconductivity of MBG and antibacterial properties of Au NPs. Upon near-infrared (NIR) irradiation, the MBG@Au NPs showed efficient antibacterial properties both in vitro and in vivo. Besides, the osteogenesis properties of MBG@Au also improved under NIR irradiation. Furthermore, the in vivo results demonstrated that MBG@Au can effectively promote alveolar bone regeneration and realize the healing of serious periodontitis.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140833577","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Compositional, Structural, and Biomechanical Properties of Three Different Soft Tissue–Hard Tissue Insertions: A Comparative Review 三种不同软组织-硬组织插入物的组成、结构和生物力学特性:比较综述
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-05-02 DOI: 10.1021/acsbiomaterials.3c01796
Nian Liu, Jialing Jiang, Tiancheng Liu, Haozhe Chen* and Nan Jiang*, 

Connective tissue attaches to bone across an insertion with spatial gradients in components, microstructure, and biomechanics. Due to regional stress concentrations between two mechanically dissimilar materials, the insertion is vulnerable to mechanical damage during joint movements and difficult to repair completely, which remains a significant clinical challenge. Despite interface stress concentrations, the native insertion physiologically functions as the effective load-transfer device between soft tissue and bone. This review summarizes tendon, ligament, and meniscus insertions cross-sectionally, which is novel in this field. Herein, the similarities and differences between the three kinds of insertions in terms of components, microstructure, and biomechanics are compared in great detail. This review begins with describing the basic components existing in the four zones (original soft tissue, uncalcified fibrocartilage, calcified fibrocartilage, and bone) of each kind of insertion, respectively. It then discusses the microstructure constructed from collagen, glycosaminoglycans (GAGs), minerals and others, which provides key support for the biomechanical properties and affects its physiological functions. Finally, the review continues by describing variations in mechanical properties at the millimeter, micrometer, and nanometer scale, which minimize stress concentrations and control stretch at the insertion. In summary, investigating the contrasts between the three has enlightening significance for future directions of repair strategies of insertion diseases and for bioinspired approaches to effective soft–hard interfaces and other tough and robust materials in medicine and engineering.

结缔组织与骨骼的连接处在成分、微结构和生物力学方面存在空间梯度。由于两种机械结构不同的材料之间存在区域性应力集中,插入部在关节运动时很容易受到机械损伤,而且很难完全修复,这仍然是一个重大的临床挑战。尽管存在界面应力集中,但原生插入部在生理上仍是软组织和骨骼之间有效的负荷传递装置。本综述对肌腱、韧带和半月板插入部位进行了横截面总结,这在该领域尚属首次。本文详细比较了三种插入物在成分、微观结构和生物力学方面的异同。本综述首先介绍了每种插入物的四个区域(原始软组织、未钙化纤维软骨、钙化纤维软骨和骨)分别存在的基本成分。然后讨论由胶原蛋白、糖胺聚糖(GAGs)、矿物质等构成的微观结构,这些微观结构为生物力学特性提供了关键支持,并影响其生理功能。最后,文章继续介绍了毫米、微米和纳米尺度的机械性能变化,这些变化可最大限度地减少应力集中并控制插入处的伸展。总之,研究这三者之间的对比,对于未来的插入疾病修复策略方向、有效的软硬界面生物启发方法以及医学和工程学中的其他坚韧材料,都具有启发性意义。
{"title":"Compositional, Structural, and Biomechanical Properties of Three Different Soft Tissue–Hard Tissue Insertions: A Comparative Review","authors":"Nian Liu,&nbsp;Jialing Jiang,&nbsp;Tiancheng Liu,&nbsp;Haozhe Chen* and Nan Jiang*,&nbsp;","doi":"10.1021/acsbiomaterials.3c01796","DOIUrl":"10.1021/acsbiomaterials.3c01796","url":null,"abstract":"<p >Connective tissue attaches to bone across an insertion with spatial gradients in components, microstructure, and biomechanics. Due to regional stress concentrations between two mechanically dissimilar materials, the insertion is vulnerable to mechanical damage during joint movements and difficult to repair completely, which remains a significant clinical challenge. Despite interface stress concentrations, the native insertion physiologically functions as the effective load-transfer device between soft tissue and bone. This review summarizes tendon, ligament, and meniscus insertions cross-sectionally, which is novel in this field. Herein, the similarities and differences between the three kinds of insertions in terms of components, microstructure, and biomechanics are compared in great detail. This review begins with describing the basic components existing in the four zones (original soft tissue, uncalcified fibrocartilage, calcified fibrocartilage, and bone) of each kind of insertion, respectively. It then discusses the microstructure constructed from collagen, glycosaminoglycans (GAGs), minerals and others, which provides key support for the biomechanical properties and affects its physiological functions. Finally, the review continues by describing variations in mechanical properties at the millimeter, micrometer, and nanometer scale, which minimize stress concentrations and control stretch at the insertion. In summary, investigating the contrasts between the three has enlightening significance for future directions of repair strategies of insertion diseases and for bioinspired approaches to effective soft–hard interfaces and other tough and robust materials in medicine and engineering.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140833445","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Moldable Tissue-Sealant Hydrogels Composed of In Situ Cross-Linkable Polyethylene Glycol via Thiol-Michael Addition and Carbomers 通过硫醇-迈克尔加成法与碳化聚合物组成的可原位交联聚乙二醇的可模塑组织密封水凝胶
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-05-02 DOI: 10.1021/acsbiomaterials.3c01755
Kento Mitsuhashi, Natsuko F. Inagaki and Taichi Ito*, 

Moldable tissue-sealant hydrogels were developed herein by combining the yield stress fluidity of a Carbomer and in situ cross-linking of 3-arm PEG-thiol (PEG-SH) and 4-arm PEG-acrylate (PEG-AC). The Carbomer was mixed with each PEG oligomer to form two aqueous precursors: Carbomer/PEG-SH and Carbomer/PEG-AC. The two hydrogel precursors exhibited sufficient yield stress (>100 Pa) to prevent dripping from their placement on the tissue surface. Moreover, these hydrogel precursors exhibited rapid restructuring when the shear strain was repeatedly changed. These rheological properties contribute to the moldability of these hydrogel precursors. After mixing these two precursors, they were converted from yield-stress fluids to chemically cross-linked hydrogels, Carbomer/PEG hydrogel, via thiol-Michael addition. The gelation time was 5.0 and 11.2 min at 37 and 25 °C, respectively. In addition, the Carbomer/PEG hydrogels exhibited higher cellular viability than the pure Carbomer. They also showed stable adhesiveness and burst pressure resistance to various tissues, such as the skin, stomach, colon, and cecum of pigs. The hydrogels showed excellent tissue sealing in a cecum ligation and puncture model in mice and improved the survival rate due to their tissue adhesiveness and biocompatibility. The Carbomer/PEG hydrogel is a potential biocompatible tissue sealant that surgeons can mold. It was revealed that the combination of in situ cross-linkable PEG oligomers and yield stress fluid such as Carbomer is effective for developing the moldable tissue sealant without dripping of its hydrogel precursors.

通过将卡波姆的屈服应力流动性与 3-臂 PEG-硫醇 (PEG-SH) 和 4-臂 PEG-丙烯酸酯 (PEG-AC) 的原位交联相结合,开发出了可模塑的组织密封水凝胶。卡波姆与每种 PEG 低聚物混合形成两种水性前体:Carbomer/PEG-SH 和 Carbomer/PEG-AC。这两种水凝胶前体具有足够的屈服应力(100 Pa),可防止放置在组织表面时滴落。此外,当剪切应变反复变化时,这些水凝胶前体表现出快速重组。这些流变特性有助于提高这些水凝胶前体的成型性。将这两种前体混合后,通过硫醇-迈克尔加成法将它们从屈服应力流体转化为化学交联水凝胶 Carbomer/PEG 水凝胶。在 37 和 25 °C 温度下,凝胶化时间分别为 5.0 和 11.2 分钟。此外,Carbomer/PEG 水凝胶比纯 Carbomer 表现出更高的细胞活力。它们对猪的皮肤、胃、结肠和盲肠等各种组织也表现出稳定的粘附性和抗爆压性。在小鼠盲肠结扎和穿刺模型中,水凝胶表现出优异的组织密封性,并因其组织粘附性和生物相容性而提高了存活率。Carbomer/PEG 水凝胶是一种潜在的生物相容性组织密封剂,外科医生可以对其进行模塑。研究表明,原位可交联 PEG 低聚物与 Carbomer 等屈服应力液的结合可有效地开发出可成型的组织密封剂,而其水凝胶前体不会滴落。
{"title":"Moldable Tissue-Sealant Hydrogels Composed of In Situ Cross-Linkable Polyethylene Glycol via Thiol-Michael Addition and Carbomers","authors":"Kento Mitsuhashi,&nbsp;Natsuko F. Inagaki and Taichi Ito*,&nbsp;","doi":"10.1021/acsbiomaterials.3c01755","DOIUrl":"10.1021/acsbiomaterials.3c01755","url":null,"abstract":"<p >Moldable tissue-sealant hydrogels were developed herein by combining the yield stress fluidity of a Carbomer and <i>in situ</i> cross-linking of 3-arm PEG-thiol (PEG-SH) and 4-arm PEG-acrylate (PEG-AC). The Carbomer was mixed with each PEG oligomer to form two aqueous precursors: Carbomer/PEG-SH and Carbomer/PEG-AC. The two hydrogel precursors exhibited sufficient yield stress (&gt;100 Pa) to prevent dripping from their placement on the tissue surface. Moreover, these hydrogel precursors exhibited rapid restructuring when the shear strain was repeatedly changed. These rheological properties contribute to the moldability of these hydrogel precursors. After mixing these two precursors, they were converted from yield-stress fluids to chemically cross-linked hydrogels, Carbomer/PEG hydrogel, via thiol-Michael addition. The gelation time was 5.0 and 11.2 min at 37 and 25 °C, respectively. In addition, the Carbomer/PEG hydrogels exhibited higher cellular viability than the pure Carbomer. They also showed stable adhesiveness and burst pressure resistance to various tissues, such as the skin, stomach, colon, and cecum of pigs. The hydrogels showed excellent tissue sealing in a cecum ligation and puncture model in mice and improved the survival rate due to their tissue adhesiveness and biocompatibility. The Carbomer/PEG hydrogel is a potential biocompatible tissue sealant that surgeons can mold. It was revealed that the combination of <i>in situ</i> cross-linkable PEG oligomers and yield stress fluid such as Carbomer is effective for developing the moldable tissue sealant without dripping of its hydrogel precursors.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140833671","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Pillar/Perfusion Plate Enhances Cell Growth, Reproducibility, Throughput, and User Friendliness in Dynamic 3D Cell Culture 柱状/灌注板提高了动态三维细胞培养中的细胞生长、可重复性、通量和用户友好性
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-05-02 DOI: 10.1021/acsbiomaterials.4c00179
Vinod Kumar Reddy Lekkala, Soo-Yeon Kang, Jiafeng Liu, Sunil Shrestha, Prabha Acharya, Pranav Joshi, Mona Zolfaghar, Minseong Lee, Manav Goud Vanga, Paarth Jamdagneya, Sohan Pagnis, Arham Kundi, Samarth Kabbur, Ung Tae Kim, Yong Yang and Moo-Yeal Lee*, 

Static three-dimensional (3D) cell culture has been demonstrated in ultralow attachment well plates, hanging droplet plates, and microtiter well plates with hydrogels or magnetic nanoparticles. Although it is simple, reproducible, and relatively inexpensive, thus potentially used for high-throughput screening, statically cultured 3D cells often suffer from a necrotic core due to limited nutrient and oxygen diffusion and waste removal and have a limited in vivo-like tissue structure. Here, we overcome these challenges by developing a pillar/perfusion plate platform and demonstrating high-throughput, dynamic 3D cell culture. Cell spheroids were loaded on the pillar plate with hydrogel by simple sandwiching and encapsulation and cultured dynamically in the perfusion plate on a digital rocker. Unlike traditional microfluidic devices, fast flow velocity was maintained within perfusion wells and the pillar plate was separated from the perfusion plate for cell-based assays. It was compatible with common lab equipment and allowed cell culture, testing, staining, and imaging in situ. The pillar/perfusion plate enhanced cell growth by rapid diffusion, reproducibility, assay throughput, and user friendliness in a dynamic 3D cell culture.

静态三维(3D)细胞培养已在超低附着孔板、悬挂液滴板以及带有水凝胶或磁性纳米颗粒的微孔板中得到证实。虽然三维细胞培养简单、可重现性好、成本相对低廉,因此有可能用于高通量筛选,但静态培养的三维细胞往往因营养和氧气扩散以及废物清除受限而出现坏死核心,而且其活体组织结构也很有限。在这里,我们通过开发一种支柱/灌注板平台来克服这些挑战,并展示了高通量、动态的三维细胞培养。通过简单的夹层和封装,细胞球体被装载到装有水凝胶的支柱板上,并在数字摇杆上的灌注板中进行动态培养。与传统的微流控装置不同,该装置可在灌注孔内保持快速流速,并可将支柱板与灌注板分离,以进行基于细胞的检测。它与普通实验室设备兼容,可进行原位细胞培养、测试、染色和成像。在动态三维细胞培养中,支柱/灌注板通过快速扩散、可重复性、检测通量和用户友好性提高了细胞生长。
{"title":"A Pillar/Perfusion Plate Enhances Cell Growth, Reproducibility, Throughput, and User Friendliness in Dynamic 3D Cell Culture","authors":"Vinod Kumar Reddy Lekkala,&nbsp;Soo-Yeon Kang,&nbsp;Jiafeng Liu,&nbsp;Sunil Shrestha,&nbsp;Prabha Acharya,&nbsp;Pranav Joshi,&nbsp;Mona Zolfaghar,&nbsp;Minseong Lee,&nbsp;Manav Goud Vanga,&nbsp;Paarth Jamdagneya,&nbsp;Sohan Pagnis,&nbsp;Arham Kundi,&nbsp;Samarth Kabbur,&nbsp;Ung Tae Kim,&nbsp;Yong Yang and Moo-Yeal Lee*,&nbsp;","doi":"10.1021/acsbiomaterials.4c00179","DOIUrl":"10.1021/acsbiomaterials.4c00179","url":null,"abstract":"<p >Static three-dimensional (3D) cell culture has been demonstrated in ultralow attachment well plates, hanging droplet plates, and microtiter well plates with hydrogels or magnetic nanoparticles. Although it is simple, reproducible, and relatively inexpensive, thus potentially used for high-throughput screening, statically cultured 3D cells often suffer from a necrotic core due to limited nutrient and oxygen diffusion and waste removal and have a limited <i>in vivo</i>-like tissue structure. Here, we overcome these challenges by developing a pillar/perfusion plate platform and demonstrating high-throughput, dynamic 3D cell culture. Cell spheroids were loaded on the pillar plate with hydrogel by simple sandwiching and encapsulation and cultured dynamically in the perfusion plate on a digital rocker. Unlike traditional microfluidic devices, fast flow velocity was maintained within perfusion wells and the pillar plate was separated from the perfusion plate for cell-based assays. It was compatible with common lab equipment and allowed cell culture, testing, staining, and imaging <i>in situ</i>. The pillar/perfusion plate enhanced cell growth by rapid diffusion, reproducibility, assay throughput, and user friendliness in a dynamic 3D cell culture.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140833259","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Preclinical Trial Protocol Using an Ovine Model to Assess Scaffold Implant Biomaterials for Repair of Critical-Sized Mandibular Defects 使用绵羊模型评估用于修复下颌骨严重缺损的支架植入生物材料的临床前试验方案
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-05-02 DOI: 10.1021/acsbiomaterials.4c00262
Hai Xin*, Ben M. Ferguson, Boyang Wan, D S Abdullah Al Maruf, William T. Lewin, Kai Cheng, Hedi V. Kruse, David Leinkram, Krishnan Parthasarathi, Innes K. Wise, Catriona Froggatt, Jeremy M. Crook, David R. McKenzie, Qing Li and Jonathan R. Clark, 

The present work describes a preclinical trial (in silico, in vivo and in vitro) protocol to assess the biomechanical performance and osteogenic capability of 3D-printed polymeric scaffolds implants used to repair partial defects in a sheep mandible. The protocol spans multiple steps of the medical device development pipeline, including initial concept design of the scaffold implant, digital twin in silico finite element modeling, manufacturing of the device prototype, in vivo device implantation, and in vitro laboratory mechanical testing. First, a patient-specific one-body scaffold implant used for reconstructing a critical-sized defect along the lower border of the sheep mandible ramus was designed using on computed-tomographic (CT) imagery and computer-aided design software. Next, the biomechanical performance of the implant was predicted numerically by simulating physiological load conditions in a digital twin in silico finite element model of the sheep mandible. This allowed for possible redesigning of the implant prior to commencing in vivo experimentation. Then, two types of polymeric biomaterials were used to manufacture the mandibular scaffold implants: poly ether ether ketone (PEEK) and poly ether ketone (PEK) printed with fused deposition modeling (FDM) and selective laser sintering (SLS), respectively. Then, after being implanted for 13 weeks in vivo, the implant and surrounding bone tissue was harvested and microCT scanned to visualize and quantify neo-tissue formation in the porous space of the scaffold. Finally, the implant and local bone tissue was assessed by in vitro laboratory mechanical testing to quantify the osteointegration. The protocol consists of six component procedures: (i) scaffold design and finite element analysis to predict its biomechanical response, (ii) scaffold fabrication with FDM and SLS 3D printing, (iii) surface treatment of the scaffold with plasma immersion ion implantation (PIII) techniques, (iv) ovine mandibular implantation, (v) postoperative sheep recovery, euthanasia, and harvesting of the scaffold and surrounding host bone, microCT scanning, and (vi) in vitro laboratory mechanical tests of the harvested scaffolds. The results of microCT imagery and 3-point mechanical bend testing demonstrate that PIII-SLS-PEK is a promising biomaterial for the manufacturing of scaffold implants to enhance the bone-scaffold contact and bone ingrowth in porous scaffold implants. MicroCT images of the harvested implant and surrounding bone tissue showed encouraging new bone growth at the scaffold-bone interface and inside the porous network of the lattice structure of the SLS-PEK scaffolds.

本研究介绍了一种临床前试验(硅学、体内和体外)方案,用于评估用于修复绵羊下颌骨部分缺损的三维打印聚合物支架植入物的生物力学性能和成骨能力。该方案跨越了医疗设备开发流程的多个步骤,包括支架植入物的初始概念设计、数字孪生硅学有限元建模、设备原型制造、体内设备植入和体外实验室机械测试。首先,使用计算机断层扫描(CT)图像和计算机辅助设计软件设计了患者专用的单体支架植入体,用于重建绵羊下颌骨横梁下缘的临界大小缺损。接下来,通过在绵羊下颌骨的数字孪生硅有限元模型中模拟生理负荷条件,对植入物的生物力学性能进行了数值预测。这样就可以在开始体内实验之前对植入体进行重新设计。然后,两种高分子生物材料被用来制造下颌骨支架植入物:聚醚醚酮(PEEK)和聚醚醚酮(PEK),分别采用熔融沉积成型(FDM)和选择性激光烧结(SLS)技术打印。然后,在体内植入 13 周后,采集植入体和周围骨组织并进行显微 CT 扫描,以观察和量化支架多孔空间中新组织的形成。最后,通过体外实验室机械测试对植入物和局部骨组织进行评估,以量化骨整合情况。该方案由六个步骤组成:(i) 支架设计和有限元分析,以预测其生物力学响应;(ii) 利用 FDM 和 SLS 3D 打印技术制作支架;(iii) 利用等离子浸入离子注入(PIII)技术对支架进行表面处理;(iv) 卵巢下颌骨植入;(v) 术后绵羊恢复、安乐死、支架和周围宿主骨的采集、显微 CT 扫描;(vi) 对采集的支架进行体外实验室力学测试。显微 CT 图像和三点机械弯曲测试的结果表明,PIII-SLS-PEK 是一种很有前途的生物材料,可用于制造支架植入物,以增强多孔支架植入物的骨-支架接触和骨生长。收获的植入体和周围骨组织的显微 CT 图像显示,在支架-骨界面以及 SLS-PEK 支架晶格结构的多孔网络内部,都有令人鼓舞的新骨生长。
{"title":"A Preclinical Trial Protocol Using an Ovine Model to Assess Scaffold Implant Biomaterials for Repair of Critical-Sized Mandibular Defects","authors":"Hai Xin*,&nbsp;Ben M. Ferguson,&nbsp;Boyang Wan,&nbsp;D S Abdullah Al Maruf,&nbsp;William T. Lewin,&nbsp;Kai Cheng,&nbsp;Hedi V. Kruse,&nbsp;David Leinkram,&nbsp;Krishnan Parthasarathi,&nbsp;Innes K. Wise,&nbsp;Catriona Froggatt,&nbsp;Jeremy M. Crook,&nbsp;David R. McKenzie,&nbsp;Qing Li and Jonathan R. Clark,&nbsp;","doi":"10.1021/acsbiomaterials.4c00262","DOIUrl":"10.1021/acsbiomaterials.4c00262","url":null,"abstract":"<p >The present work describes a preclinical trial (<i>in silico</i>, <i>in vivo</i> and <i>in vitro</i>) protocol to assess the biomechanical performance and osteogenic capability of 3D-printed polymeric scaffolds implants used to repair partial defects in a sheep mandible. The protocol spans multiple steps of the medical device development pipeline, including initial concept design of the scaffold implant, digital twin <i>in silico</i> finite element modeling, manufacturing of the device prototype, <i>in vivo</i> device implantation, and <i>in vitro</i> laboratory mechanical testing. First, a patient-specific one-body scaffold implant used for reconstructing a critical-sized defect along the lower border of the sheep mandible ramus was designed using on computed-tomographic (CT) imagery and computer-aided design software. Next, the biomechanical performance of the implant was predicted numerically by simulating physiological load conditions in a digital twin <i>in silico</i> finite element model of the sheep mandible. This allowed for possible redesigning of the implant prior to commencing <i>in vivo</i> experimentation. Then, two types of polymeric biomaterials were used to manufacture the mandibular scaffold implants: poly ether ether ketone (PEEK) and poly ether ketone (PEK) printed with fused deposition modeling (FDM) and selective laser sintering (SLS), respectively. Then, after being implanted for 13 weeks <i>in</i> vivo, the implant and surrounding bone tissue was harvested and microCT scanned to visualize and quantify neo-tissue formation in the porous space of the scaffold. Finally, the implant and local bone tissue was assessed by <i>in vitro</i> laboratory mechanical testing to quantify the osteointegration. The protocol consists of six component procedures: (i) scaffold design and finite element analysis to predict its biomechanical response, (ii) scaffold fabrication with FDM and SLS 3D printing, (iii) surface treatment of the scaffold with plasma immersion ion implantation (PIII) techniques, (iv) ovine mandibular implantation, (v) postoperative sheep recovery, euthanasia, and harvesting of the scaffold and surrounding host bone, microCT scanning, and (vi) <i>in vitro</i> laboratory mechanical tests of the harvested scaffolds. The results of microCT imagery and 3-point mechanical bend testing demonstrate that PIII-SLS-PEK is a promising biomaterial for the manufacturing of scaffold implants to enhance the bone-scaffold contact and bone ingrowth in porous scaffold implants. MicroCT images of the harvested implant and surrounding bone tissue showed encouraging new bone growth at the scaffold-bone interface and inside the porous network of the lattice structure of the SLS-PEK scaffolds.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140833263","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Silk-Based 3D Porous Scaffolds for Tissue Engineering 用于组织工程的蚕丝基三维多孔支架
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-05-01 DOI: 10.1021/acsbiomaterials.4c00373
Menglin Xiao, Jinrong Yao, Zhengzhong Shao and Xin Chen*, 

Silk fibroin, extracted from the silk of the Bombyx mori silkworm, stands out as a biomaterial due to its nontoxic nature, excellent biocompatibility, and adjustable biodegradability. Porous scaffolds, a type of biomaterial, are crucial for creating an optimal microenvironment that supports cell adhesion and proliferation, thereby playing an essential role in tissue remodeling and repair. Therefore, this review focuses on 3D porous silk fibroin-based scaffolds, first summarizing their preparation methods and then detailing their regenerative effects on bone, cartilage, tendon, vascular, neural, skin, hepatic, and tracheal epithelial tissue engineering in recent years.

蚕丝纤维素提取自家蚕的蚕丝,因其无毒性、良好的生物相容性和可调节的生物降解性而成为生物材料中的佼佼者。多孔支架是生物材料的一种,对于创造支持细胞粘附和增殖的最佳微环境至关重要,因此在组织重塑和修复中发挥着至关重要的作用。因此,本综述将重点关注基于丝纤维素的三维多孔支架,首先总结其制备方法,然后详细介绍近年来其在骨骼、软骨、肌腱、血管、神经、皮肤、肝脏和气管上皮组织工程中的再生效果。
{"title":"Silk-Based 3D Porous Scaffolds for Tissue Engineering","authors":"Menglin Xiao,&nbsp;Jinrong Yao,&nbsp;Zhengzhong Shao and Xin Chen*,&nbsp;","doi":"10.1021/acsbiomaterials.4c00373","DOIUrl":"10.1021/acsbiomaterials.4c00373","url":null,"abstract":"<p >Silk fibroin, extracted from the silk of the <i>Bombyx mori</i> silkworm, stands out as a biomaterial due to its nontoxic nature, excellent biocompatibility, and adjustable biodegradability. Porous scaffolds, a type of biomaterial, are crucial for creating an optimal microenvironment that supports cell adhesion and proliferation, thereby playing an essential role in tissue remodeling and repair. Therefore, this review focuses on 3D porous silk fibroin-based scaffolds, first summarizing their preparation methods and then detailing their regenerative effects on bone, cartilage, tendon, vascular, neural, skin, hepatic, and tracheal epithelial tissue engineering in recent years.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140833616","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel Nonthermal Atmospheric Plasma Irradiation of Titanium Implants Promotes Osteogenic Effect in Osteoporotic Conditions 新型非热大气等离子体辐照钛植入物可促进骨质疏松情况下的成骨效应
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-04-29 DOI: 10.1021/acsbiomaterials.4c00202
Yihan Liao, Jia Xu, Zheng Zheng, Ruijie Fu, Xinyuan Zhang, Shuaiqi Gan, Shuhan Yang, Chuping Hou, Hockin H. K. Xu and Wenchuan Chen*, 

Osteoporosis is a metabolic disease characterized by bone density and trabecular bone loss. Bone loss may affect dental implant osseointegration in patients with osteoporosis. To promote implant osseointegration in osteoporotic patients, we further used a nonthermal atmospheric plasma (NTAP) treatment device previously developed by our research group. After the titanium implant (Ti) is placed into the device, the working gas flow and the electrode switches are turned on, and the treatment is completed in 30 s. Previous studies showed that this NTAP device can remove carbon contamination from the implant surface, increase the hydroxyl groups, and improve its wettability to promote osseointegration in normal conditions. In this study, we demonstrated the tremendous osteogenic enhancement effect of NTAP-Ti in osteoporotic conditions in rats for the first time. Compared to Ti, the proliferative potential of osteoporotic bone marrow mesenchymal stem cells on NTAP-Ti increased by 180% at 1 day (P = 0.004), while their osteogenic differentiation increased by 149% at 14 days (P < 0.001). In addition, the results indicated that NTAP-Ti significantly improved osseointegration in osteoporotic rats in vivo. Compared to the Ti, the bone volume fraction (BV/TV) and trabecular number (Tb.N) values of NTAP-Ti in osteoporotic rats, respectively, increased by 18% (P < 0.001) and 25% (P = 0.007) at 6 weeks and the trabecular separation (Tb.Sp) value decreased by 26% (P = 0.02) at 6 weeks. In conclusion, this study proved a novel NTAP irradiation titanium implant that can significantly promote osseointegration in osteoporotic conditions.

骨质疏松症是一种以骨密度和骨小梁丢失为特征的代谢性疾病。骨质流失可能会影响骨质疏松症患者种植牙的骨结合。为了促进骨质疏松症患者的种植体骨结合,我们进一步使用了本研究小组之前开发的非热大气等离子体(NTAP)处理装置。之前的研究表明,这种 NTAP 设备可以去除种植体表面的碳污染,增加羟基,改善其润湿性,从而促进正常情况下的骨结合。在本研究中,我们首次证明了 NTAP-Ti 对骨质疏松大鼠的巨大成骨增强作用。与钛相比,骨质疏松骨髓间充质干细胞在 NTAP-Ti 上的增殖潜力在 1 天时增加了 180%(P = 0.004),而其成骨分化在 14 天时增加了 149%(P < 0.001)。此外,研究结果表明,NTAP-Ti 能明显改善骨质疏松大鼠体内的骨结合。与钛相比,NTAP-钛在骨质疏松大鼠体内的骨体积分数(BV/TV)和骨小梁数(Tb.N)值在 6 周时分别增加了 18% (P < 0.001) 和 25% (P = 0.007),骨小梁分离度(Tb.Sp)值在 6 周时降低了 26% (P = 0.02)。总之,本研究证明了一种新型的 NTAP 照射钛植入物可显著促进骨质疏松症患者的骨结合。
{"title":"Novel Nonthermal Atmospheric Plasma Irradiation of Titanium Implants Promotes Osteogenic Effect in Osteoporotic Conditions","authors":"Yihan Liao,&nbsp;Jia Xu,&nbsp;Zheng Zheng,&nbsp;Ruijie Fu,&nbsp;Xinyuan Zhang,&nbsp;Shuaiqi Gan,&nbsp;Shuhan Yang,&nbsp;Chuping Hou,&nbsp;Hockin H. K. Xu and Wenchuan Chen*,&nbsp;","doi":"10.1021/acsbiomaterials.4c00202","DOIUrl":"10.1021/acsbiomaterials.4c00202","url":null,"abstract":"<p >Osteoporosis is a metabolic disease characterized by bone density and trabecular bone loss. Bone loss may affect dental implant osseointegration in patients with osteoporosis. To promote implant osseointegration in osteoporotic patients, we further used a nonthermal atmospheric plasma (NTAP) treatment device previously developed by our research group. After the titanium implant (Ti) is placed into the device, the working gas flow and the electrode switches are turned on, and the treatment is completed in 30 s. Previous studies showed that this NTAP device can remove carbon contamination from the implant surface, increase the hydroxyl groups, and improve its wettability to promote osseointegration in normal conditions. In this study, we demonstrated the tremendous osteogenic enhancement effect of NTAP-Ti in osteoporotic conditions in rats for the first time. Compared to Ti, the proliferative potential of osteoporotic bone marrow mesenchymal stem cells on NTAP-Ti increased by 180% at 1 day (<i>P</i> = 0.004), while their osteogenic differentiation increased by 149% at 14 days (<i>P</i> &lt; 0.001). In addition, the results indicated that NTAP-Ti significantly improved osseointegration in osteoporotic rats in vivo. Compared to the Ti, the bone volume fraction (BV/TV) and trabecular number (Tb.N) values of NTAP-Ti in osteoporotic rats, respectively, increased by 18% (<i>P</i> &lt; 0.001) and 25% (<i>P</i> = 0.007) at 6 weeks and the trabecular separation (Tb.Sp) value decreased by 26% (<i>P</i> = 0.02) at 6 weeks. In conclusion, this study proved a novel NTAP irradiation titanium implant that can significantly promote osseointegration in osteoporotic conditions.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140833256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel In Vitro Platform for Studying the Cell Response to Healthy and Diseased Tendon Matrices 研究细胞对健康和患病肌腱基质反应的新型体外平台
IF 5.8 2区 医学 Q1 Engineering Pub Date : 2024-04-26 DOI: 10.1021/acsbiomaterials.4c00414
Subhajit Konar, Sophia Leung, Mei Lin Tay, Brendan Coleman, Nicola Dalbeth, Jillian Cornish, Dorit Naot and David S. Musson*, 

Current in vitro models poorly represent the healthy or diseased tendon microenvironment, limiting the translation of the findings to clinics. The present work aims to establish a physiologically relevant in vitro tendon platform that mimics biophysical aspects of a healthy and tendinopathic tendon matrix using a decellularized bovine tendon and to characterize tendon cells cultured using this platform. Bovine tendons were subjected to various decellularization techniques, with the efficacy of decellularization determined histologically. The biomechanical and architectural properties of the decellularized tendons were characterized using an atomic force microscope. Tendinopathy-mimicking matrices were prepared by treating the decellularized tendons with collagenase for 3 h or collagenase–chondroitinase (CC) for 1 h. The tendon tissue collected from healthy and tendinopathic patients was characterized using an atomic force microscope and compared to that of decellularized matrices. Healthy human tendon-derived cells (hTDCs) from the hamstring tendon were cultured on the decellularized matrices for 24 or 48 h, with cell morphology characterized using f-actin staining and gene expression characterized using real-time PCR. Tendon matrices prepared by freeze–thawing and 48 h nuclease treatment were fully decellularized, and the aligned structure and tendon stiffness (1.46 MPa) were maintained. Collagenase treatment prepared matrices with a disorganized architecture and reduced stiffness (0.75 MPa), mimicking chronic tendinopathy. Treatment with CC prepared matrices with a disorganized architecture without altering stiffness, mimicking early tendinopathy (1.52 MPa). hTDCs on a healthy tendon matrix were elongated, and the scleraxis (SCX) expression was maintained. On tendinopathic matrices, hTDCs had altered morphological characteristics and lower SCX expression. The expression of genes related to actin polymerization, matrix degradation and remodeling, and immune cell invasion were higher in hTDCs on tendinopathic matrices. Overall, the present study developed a physiological in vitro system to mimic healthy tendons and early and late tendinopathy, and it can be used to better understand tendon cell characteristics in healthy and diseased states.

目前的体外模型不能很好地代表健康或患病肌腱的微环境,从而限制了将研究结果应用于临床。本研究旨在建立一个与生理相关的体外肌腱平台,利用脱细胞牛肌腱模拟健康和肌腱病变肌腱基质的生物物理方面,并描述利用该平台培养的肌腱细胞的特征。牛肌腱采用了各种脱细胞技术,并通过组织学方法确定了脱细胞的效果。使用原子力显微镜鉴定了脱细胞肌腱的生物力学和结构特性。用胶原蛋白酶处理脱细胞肌腱3小时或用胶原蛋白酶-软骨素酶(CC)处理脱细胞肌腱1小时,制备出肌腱病模拟基质。利用原子力显微镜对从健康人和肌腱病患者身上采集的肌腱组织进行表征,并与脱细胞基质进行比较。来自腘绳肌腱的健康人肌腱衍生细胞(hTDCs)在脱细胞基质上培养 24 或 48 小时,细胞形态用 f-肌动蛋白染色法表征,基因表达用实时 PCR 法表征。通过冻融和核酸酶处理 48 小时制备的肌腱基质完全脱细胞,并保持了排列整齐的结构和肌腱硬度(1.46 兆帕)。胶原酶处理制备的基质结构混乱,硬度降低(0.75 兆帕),模拟了慢性肌腱病。健康肌腱基质上的 hTDC 拉长,硬轴(SCX)表达保持不变。在肌腱病变基质上,hTDC的形态特征发生改变,SCX表达较低。在腱鞘病变基质上,hTDCs 的肌动蛋白聚合、基质降解和重塑以及免疫细胞侵袭相关基因的表达较高。总之,本研究开发了一种生理体外系统来模拟健康肌腱以及早期和晚期肌腱病变,可用于更好地了解健康和疾病状态下肌腱细胞的特征。
{"title":"Novel In Vitro Platform for Studying the Cell Response to Healthy and Diseased Tendon Matrices","authors":"Subhajit Konar,&nbsp;Sophia Leung,&nbsp;Mei Lin Tay,&nbsp;Brendan Coleman,&nbsp;Nicola Dalbeth,&nbsp;Jillian Cornish,&nbsp;Dorit Naot and David S. Musson*,&nbsp;","doi":"10.1021/acsbiomaterials.4c00414","DOIUrl":"10.1021/acsbiomaterials.4c00414","url":null,"abstract":"<p >Current in vitro models poorly represent the healthy or diseased tendon microenvironment, limiting the translation of the findings to clinics. The present work aims to establish a physiologically relevant in vitro tendon platform that mimics biophysical aspects of a healthy and tendinopathic tendon matrix using a decellularized bovine tendon and to characterize tendon cells cultured using this platform. Bovine tendons were subjected to various decellularization techniques, with the efficacy of decellularization determined histologically. The biomechanical and architectural properties of the decellularized tendons were characterized using an atomic force microscope. Tendinopathy-mimicking matrices were prepared by treating the decellularized tendons with collagenase for 3 h or collagenase–chondroitinase (CC) for 1 h. The tendon tissue collected from healthy and tendinopathic patients was characterized using an atomic force microscope and compared to that of decellularized matrices. Healthy human tendon-derived cells (hTDCs) from the hamstring tendon were cultured on the decellularized matrices for 24 or 48 h, with cell morphology characterized using f-actin staining and gene expression characterized using real-time PCR. Tendon matrices prepared by freeze–thawing and 48 h nuclease treatment were fully decellularized, and the aligned structure and tendon stiffness (1.46 MPa) were maintained. Collagenase treatment prepared matrices with a disorganized architecture and reduced stiffness (0.75 MPa), mimicking chronic tendinopathy. Treatment with CC prepared matrices with a disorganized architecture without altering stiffness, mimicking early tendinopathy (1.52 MPa). hTDCs on a healthy tendon matrix were elongated, and the scleraxis (<i>SCX</i>) expression was maintained. On tendinopathic matrices, hTDCs had altered morphological characteristics and lower <i>SCX</i> expression. The expression of genes related to actin polymerization, matrix degradation and remodeling, and immune cell invasion were higher in hTDCs on tendinopathic matrices. Overall, the present study developed a physiological in vitro system to mimic healthy tendons and early and late tendinopathy, and it can be used to better understand tendon cell characteristics in healthy and diseased states.</p>","PeriodicalId":8,"journal":{"name":"ACS Biomaterials Science & Engineering","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140652821","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
ACS Biomaterials Science & Engineering
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
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
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