首页 > 最新文献

Biomaterials and biosystems最新文献

英文 中文
Tribological loading of cartilage and chondrogenic cells 软骨和软骨细胞的摩擦载荷
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-13 DOI: 10.1016/j.bbiosy.2024.100088
Yann D Ladner, Martin J. Stoddart

Novel cartilage regeneration therapies often look promising in-vitro but fail when implanted in vivo. One of the possible reasons for this discrepancy is the simplified, static in-vitro chondrogenesis models typically used. Complex mechanical stimulation plays a key role in physiological cartilage and chondrogenic cell metabolism, including the development of cartilage structure, yet it is routinely lacking during in-vitro studies. Multiaxial load bioreactors are becoming more widespread and offer advantages over more simple loading devices. Within this article, we highlight some of the important findings from in-vitro assays and key aspects relating to tribological loading of cartilage and chondrogenic cells.

新型软骨再生疗法往往在体外看起来很有希望,但在体内植入时却会失败。造成这种差异的原因之一可能是通常使用的简化、静态体外软骨生成模型。复杂的机械刺激在软骨和软骨细胞的生理性新陈代谢(包括软骨结构的发展)中起着关键作用,但体外研究中通常缺乏这种刺激。多轴加载生物反应器正变得越来越普遍,与更简单的加载设备相比,它具有更多优势。在本文中,我们将重点介绍体外试验的一些重要发现,以及与软骨和软骨细胞摩擦加载有关的关键方面。
{"title":"Tribological loading of cartilage and chondrogenic cells","authors":"Yann D Ladner,&nbsp;Martin J. Stoddart","doi":"10.1016/j.bbiosy.2024.100088","DOIUrl":"https://doi.org/10.1016/j.bbiosy.2024.100088","url":null,"abstract":"<div><p>Novel cartilage regeneration therapies often look promising <em>in-vitro</em> but fail when implanted <em>in vivo.</em> One of the possible reasons for this discrepancy is the simplified, static <em>in-vitro</em> chondrogenesis models typically used. Complex mechanical stimulation plays a key role in physiological cartilage and chondrogenic cell metabolism, including the development of cartilage structure, yet it is routinely lacking during <em>in-vitro</em> studies. Multiaxial load bioreactors are becoming more widespread and offer advantages over more simple loading devices. Within this article, we highlight some of the important findings from <em>in-vitro</em> assays and key aspects relating to tribological loading of cartilage and chondrogenic cells.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2666534424000023/pdfft?md5=922f992e1af45e86fbcd854c7a5bc649&pid=1-s2.0-S2666534424000023-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139744174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
3D printed hybrid scaffolds do not induce adverse inflammation in mice and direct human BM-MSC chondrogenesis in vitro 三维打印混合支架不会诱发小鼠不良炎症,并能在体外引导人类骨髓间充质干细胞软骨生成
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-08 DOI: 10.1016/j.bbiosy.2024.100087
Silvia A. Ferreira , Francesca Tallia , Agathe Heyraud , Simone A. Walker , Christoph Salzlechner , Julian R. Jones , Sara M. Rankin

Biomaterials that can improve the healing of articular cartilage lesions are needed. To address this unmet need, we developed novel 3D printed silica/poly(tetrahydrofuran)/poly(ε-caprolactone) (SiO2/PTHF/PCL-diCOOH) hybrid scaffolds. Our aim was to carry out essential studies to advance this medical device towards functional validation in pre-clinical trials. First, we show that the chemical composition, microarchitecture and mechanical properties of these scaffolds were not affected by sterilisation with gamma irradiation. To evaluate the systemic and local immunogenic reactivity of the sterilised 3D printed hybrid scaffolds, they were implanted subcutaneously into Balb/c mice. The scaffolds did not trigger a systemic inflammatory response over one week of implantation. The interaction between the host immune system and the implanted scaffold elicited a local physiological reaction with infiltration of mononuclear cells without any signs of a chronic inflammatory response.

Then, we investigated how these 3D printed hybrid scaffolds direct chondrogenesis in vitro. Human bone marrow-derived mesenchymal stem/stromal cells (hBM-MSCs) seeded within the 3D printed hybrid scaffolds were cultured under normoxic or hypoxic conditions, with or without chondrogenic supplements. Chondrogenic differentiation assessed by both gene expression and protein production analyses showed that 3D printed hybrid scaffolds support hBM-MSC chondrogenesis. Articular cartilage-specific extracellular matrix deposition within these scaffolds was enhanced under hypoxic conditions (1.7 or 3.7 fold increase in the median of aggrecan production in basal or chondrogenic differentiation media).

Our findings show that 3D printed SiO2/PTHF/PCL-diCOOH hybrid scaffolds have the potential to support the regeneration of cartilage tissue.

我们需要能改善关节软骨损伤愈合的生物材料。为了满足这一尚未满足的需求,我们开发了新型三维打印二氧化硅/聚(四氢呋喃)/聚(ε-己内酰胺)(SiO2/PTHF/PCL-diCOOH)混合支架。我们的目标是开展必要的研究,推动这一医疗设备在临床前试验中进行功能验证。首先,我们发现这些支架的化学成分、微结构和机械性能不受伽马射线灭菌的影响。为了评估灭菌三维打印混合支架的全身和局部免疫原反应性,我们将其皮下植入 Balb/c 小鼠体内。植入一周后,支架没有引发全身炎症反应。宿主免疫系统与植入支架之间的相互作用引起了局部生理反应,单核细胞浸润,但没有任何慢性炎症反应的迹象。我们将人骨髓间充质干/基质细胞(hBM-MSCs)播种到三维打印混合支架中,在常氧或缺氧条件下,添加或不添加软骨生成补充剂进行培养。通过基因表达和蛋白质生成分析评估的软骨分化结果表明,三维打印混合支架支持hBM-间充质干细胞的软骨形成。我们的研究结果表明,三维打印的 SiO2/PTHF/PCL-diCOOH 混合支架具有支持软骨组织再生的潜力。
{"title":"3D printed hybrid scaffolds do not induce adverse inflammation in mice and direct human BM-MSC chondrogenesis in vitro","authors":"Silvia A. Ferreira ,&nbsp;Francesca Tallia ,&nbsp;Agathe Heyraud ,&nbsp;Simone A. Walker ,&nbsp;Christoph Salzlechner ,&nbsp;Julian R. Jones ,&nbsp;Sara M. Rankin","doi":"10.1016/j.bbiosy.2024.100087","DOIUrl":"10.1016/j.bbiosy.2024.100087","url":null,"abstract":"<div><p>Biomaterials that can improve the healing of articular cartilage lesions are needed. To address this unmet need, we developed novel 3D printed silica/poly(tetrahydrofuran)/poly(ε-caprolactone) (SiO<sub>2</sub>/PTHF/PCL-diCOOH) hybrid scaffolds. Our aim was to carry out essential studies to advance this medical device towards functional validation in pre-clinical trials. First, we show that the chemical composition, microarchitecture and mechanical properties of these scaffolds were not affected by sterilisation with gamma irradiation. To evaluate the systemic and local immunogenic reactivity of the sterilised 3D printed hybrid scaffolds, they were implanted subcutaneously into Balb/c mice. The scaffolds did not trigger a systemic inflammatory response over one week of implantation. The interaction between the host immune system and the implanted scaffold elicited a local physiological reaction with infiltration of mononuclear cells without any signs of a chronic inflammatory response.</p><p>Then, we investigated how these 3D printed hybrid scaffolds direct chondrogenesis <em>in vitro</em>. Human bone marrow-derived mesenchymal stem/stromal cells (hBM-MSCs) seeded within the 3D printed hybrid scaffolds were cultured under normoxic or hypoxic conditions, with or without chondrogenic supplements. Chondrogenic differentiation assessed by both gene expression and protein production analyses showed that 3D printed hybrid scaffolds support hBM-MSC chondrogenesis<em>.</em> Articular cartilage-specific extracellular matrix deposition within these scaffolds was enhanced under hypoxic conditions (1.7 or 3.7 fold increase in the median of aggrecan production in basal or chondrogenic differentiation media).</p><p>Our findings show that 3D printed SiO<sub>2</sub>/PTHF/PCL-diCOOH hybrid scaffolds have the potential to support the regeneration of cartilage tissue.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2666534424000011/pdfft?md5=b41b005e94cd429e5efe1d7a4e7ffc1c&pid=1-s2.0-S2666534424000011-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139455737","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
3D printed bioabsorbable composite scaffolds of poly (lactic acid)-tricalcium phosphate-ceria with osteogenic property for bone regeneration 用于骨再生的具有成骨特性的聚乳酸-磷酸三钙-纤维素三维打印生物可吸收复合支架
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-12-18 DOI: 10.1016/j.bbiosy.2023.100086
Samarah V. Harb , Elayaraja Kolanthai , Abinaya S. Pugazhendhi , Cesar A.G. Beatrice , Leonardo A. Pinto , Craig J. Neal , Eduardo H. Backes , Ana C.C. Nunes , Heloisa S. Selistre-de-Araújo , Lidiane C. Costa , Melanie J. Coathup , Sudipta Seal , Luiz A. Pessan

The fabrication of customized implants by additive manufacturing has allowed continued development of the personalized medicine field. Herein, a 3D-printed bioabsorbable poly (lactic acid) (PLA)- β-tricalcium phosphate (TCP) (10 wt %) composite has been modified with CeO2 nanoparticles (CeNPs) (1, 5 and 10 wt %) for bone repair. The filaments were prepared by melt extrusion and used to print porous scaffolds. The nanocomposite scaffolds possessed precise structure with fine print resolution, a homogenous distribution of TCP and CeNP components, and mechanical properties appropriate for bone tissue engineering applications. Cell proliferation assays using osteoblast cultures confirmed the cytocompatibility of the composites. In addition, the presence of CeNPs enhanced the proliferation and differentiation of mesenchymal stem cells; thereby, increasing alkaline phosphatase (ALP) activity, calcium deposition and bone-related gene expression. Results from this study have shown that the 3D printed PLA-TCP-10%CeO2 composite scaffold could be used as an alternative polymeric implant for bone tissue engineering applications: avoiding additional/revision surgeries and accelerating the regenerative process.

通过增材制造技术制造定制植入物使个性化医疗领域得以持续发展。在本文中,一种三维打印的生物可吸收聚乳酸(PLA)-β-磷酸三钙(TCP)(10 wt %)复合材料被二氧化硒纳米颗粒(CeNPs)(1、5 和 10 wt %)修饰,用于骨修复。长丝通过熔融挤压制备,并用于打印多孔支架。纳米复合材料支架具有精确的结构和精细的打印分辨率、TCP 和 CeNP 成分的均匀分布以及适合骨组织工程应用的机械性能。使用成骨细胞进行的细胞增殖试验证实了复合材料的细胞相容性。此外,CeNPs 的存在增强了间充质干细胞的增殖和分化,从而提高了碱性磷酸酶 (ALP) 活性、钙沉积和骨相关基因的表达。这项研究的结果表明,3D 打印聚乳酸-TCP-10%CeO2 复合支架可用作骨组织工程应用的替代聚合物植入物:避免额外/翻修手术,加速再生过程。
{"title":"3D printed bioabsorbable composite scaffolds of poly (lactic acid)-tricalcium phosphate-ceria with osteogenic property for bone regeneration","authors":"Samarah V. Harb ,&nbsp;Elayaraja Kolanthai ,&nbsp;Abinaya S. Pugazhendhi ,&nbsp;Cesar A.G. Beatrice ,&nbsp;Leonardo A. Pinto ,&nbsp;Craig J. Neal ,&nbsp;Eduardo H. Backes ,&nbsp;Ana C.C. Nunes ,&nbsp;Heloisa S. Selistre-de-Araújo ,&nbsp;Lidiane C. Costa ,&nbsp;Melanie J. Coathup ,&nbsp;Sudipta Seal ,&nbsp;Luiz A. Pessan","doi":"10.1016/j.bbiosy.2023.100086","DOIUrl":"https://doi.org/10.1016/j.bbiosy.2023.100086","url":null,"abstract":"<div><p>The fabrication of customized implants by additive manufacturing has allowed continued development of the personalized medicine field. Herein, a 3D-printed bioabsorbable poly (lactic acid) (PLA)- β-tricalcium phosphate (TCP) (10 wt %) composite has been modified with CeO<sub>2</sub> nanoparticles (CeNPs) (1, 5 and 10 wt %) for bone repair. The filaments were prepared by melt extrusion and used to print porous scaffolds. The nanocomposite scaffolds possessed precise structure with fine print resolution, a homogenous distribution of TCP and CeNP components, and mechanical properties appropriate for bone tissue engineering applications. Cell proliferation assays using osteoblast cultures confirmed the cytocompatibility of the composites. In addition, the presence of CeNPs enhanced the proliferation and differentiation of mesenchymal stem cells; thereby, increasing alkaline phosphatase (ALP) activity, calcium deposition and bone-related gene expression. Results from this study have shown that the 3D printed PLA-TCP-10%CeO<sub>2</sub> composite scaffold could be used as an alternative polymeric implant for bone tissue engineering applications: avoiding additional/revision surgeries and accelerating the regenerative process.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2666534423000156/pdfft?md5=9dcdfda4ed3614a4d05a8b4573afea9a&pid=1-s2.0-S2666534423000156-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138839709","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rise of tissue- and species-specific 3D bioprinting based on decellularized extracellular matrix-derived bioinks and bioresins 基于脱细胞细胞外基质衍生的生物墨水和生物树脂的组织和物种特异性3D生物打印的兴起
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-11-07 DOI: 10.1016/j.bbiosy.2023.100084
Laura Elomaa , Ahed Almalla , Eriselda Keshi , Karl H. Hillebrandt , Igor M. Sauer , Marie Weinhart

Thanks to its natural complexity and functionality, decellularized extracellular matrix (dECM) serves as an excellent foundation for creating highly cell-compatible bioinks and bioresins. This enables the bioprinted cells to thrive in an environment that closely mimics their native ECM composition and offers customizable biomechanical properties. To formulate dECM bioinks and bioresins, one must first pulverize and/or solubilize the dECM into non-crosslinked fragments, which can then be chemically modified as needed. In bioprinting, the solubilized dECM-derived material is typically deposited and/or crosslinked in a layer-by-layer fashion to build 3D hydrogel structures. Since the introduction of the first liver-derived dECM-based bioinks, a wide variety of decellularized tissue have been employed in bioprinting, including kidney, heart, cartilage, and adipose tissue among others. This review aims to summarize the critical steps involved in tissue-derived dECM bioprinting, starting from the decellularization of the ECM to the standardized formulation of bioinks and bioresins, ultimately leading to the reproducible bioprinting of tissue constructs. Notably, this discussion also covers photocrosslinkable dECM bioresins, which are particularly attractive due to their ability to provide precise spatiotemporal control over the gelation in bioprinting. Both in extrusion printing and vat photopolymerization, there is a need for more standardized protocols to fully harness the unique properties of dECM-derived materials. In addition to mammalian tissues, the most recent bioprinting approaches involve the use of microbial extracellular polymeric substances in bioprinting of bacteria. This presents similar challenges as those encountered in mammalian cell printing and represents a fascinating frontier in bioprinting technology.

由于其天然的复杂性和功能性,脱细胞细胞外基质(dECM)是创造高度细胞相容性的生物墨水和生物树脂的良好基础。这使得生物打印的细胞能够在接近模拟其天然ECM组成的环境中茁壮成长,并提供可定制的生物力学特性。为了制备dECM生物墨水和生物树脂,必须首先将dECM粉碎和/或溶解成非交联碎片,然后根据需要对其进行化学修饰。在生物打印中,溶解的decm衍生材料通常以一层接一层的方式沉积和/或交联,以构建3D水凝胶结构。自从第一个肝脏来源的基于decm的生物墨水问世以来,各种各样的脱细胞组织已经被用于生物打印,包括肾脏、心脏、软骨和脂肪组织等。本综述旨在总结组织源性dECM生物打印的关键步骤,从ECM的脱细胞到生物墨水和生物树脂的标准化配方,最终导致组织结构的可复制生物打印。值得注意的是,本讨论还涵盖了光交联的dECM生物树脂,由于它们能够在生物打印中提供对凝胶化的精确时空控制,因此特别有吸引力。无论是挤压印刷还是还原光聚合,都需要更标准化的协议来充分利用decm衍生材料的独特性能。除了哺乳动物组织外,最近的生物打印方法还包括在细菌生物打印中使用微生物细胞外聚合物物质。这提出了与哺乳动物细胞打印遇到的类似挑战,代表了生物打印技术的一个迷人的前沿。
{"title":"Rise of tissue- and species-specific 3D bioprinting based on decellularized extracellular matrix-derived bioinks and bioresins","authors":"Laura Elomaa ,&nbsp;Ahed Almalla ,&nbsp;Eriselda Keshi ,&nbsp;Karl H. Hillebrandt ,&nbsp;Igor M. Sauer ,&nbsp;Marie Weinhart","doi":"10.1016/j.bbiosy.2023.100084","DOIUrl":"https://doi.org/10.1016/j.bbiosy.2023.100084","url":null,"abstract":"<div><p>Thanks to its natural complexity and functionality, decellularized extracellular matrix (dECM) serves as an excellent foundation for creating highly cell-compatible bioinks and bioresins. This enables the bioprinted cells to thrive in an environment that closely mimics their native ECM composition and offers customizable biomechanical properties. To formulate dECM bioinks and bioresins, one must first pulverize and/or solubilize the dECM into non-crosslinked fragments, which can then be chemically modified as needed. In bioprinting, the solubilized dECM-derived material is typically deposited and/or crosslinked in a layer-by-layer fashion to build 3D hydrogel structures. Since the introduction of the first liver-derived dECM-based bioinks, a wide variety of decellularized tissue have been employed in bioprinting, including kidney, heart, cartilage, and adipose tissue among others. This review aims to summarize the critical steps involved in tissue-derived dECM bioprinting, starting from the decellularization of the ECM to the standardized formulation of bioinks and bioresins, ultimately leading to the reproducible bioprinting of tissue constructs. Notably, this discussion also covers photocrosslinkable dECM bioresins, which are particularly attractive due to their ability to provide precise spatiotemporal control over the gelation in bioprinting. Both in extrusion printing and vat photopolymerization, there is a need for more standardized protocols to fully harness the unique properties of dECM-derived materials. In addition to mammalian tissues, the most recent bioprinting approaches involve the use of microbial extracellular polymeric substances in bioprinting of bacteria. This presents similar challenges as those encountered in mammalian cell printing and represents a fascinating frontier in bioprinting technology.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2666534423000132/pdfft?md5=53266b4687d1013a16fc02ee5ddb1502&pid=1-s2.0-S2666534423000132-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"134655545","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Optimization and evaluation of oxygen-plasma-modified, aligned, poly (Є-caprolactone) and silk fibroin nanofibrous scaffold for corneal stromal regeneration 氧等离子修饰、排列、聚(Є-caprolactone)和丝素纳米纤维支架用于角膜基质再生的优化与评价
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-09-03 DOI: 10.1016/j.bbiosy.2023.100083
Promita Bhattacharjee , Peter W. Madden , Enzo Patriarca , Mark Ahearne

The shortage of human donor corneas for transplantation necessitates the exploration of tissue engineering approaches to develop corneal substitutes. However, these substitutes must possess the necessary strength, transparency, and ability to regulate cell behaviour before they can be used in patients. In this study, we investigated the effectiveness of an oxygen plasma surface-modified poly-ε-caprolactone (PCL) combined with silk fibroin (SF) nanofibrous scaffold for corneal stromal regeneration. To fabricate the electrospun scaffolds, PCL and SF blends were used on a rotating mandrel. The optimization of the blend aimed to replicate the structural and functional properties of the human cornea, focusing on nanofibre alignment, mechanical characteristics, and in vitro cytocompatibility with human corneal stromal keratocytes. Surface modification of the scaffold resulted in improved transparency and enhanced cell interaction. Based on the evaluation, a composite nanofibrous scaffold with a 1:1 blend of PCL and SF was selected for a more comprehensive analysis. The biological response of keratocytes to the scaffold was assessed through cellular adhesion, proliferation, cytoskeletal organization, gene expression, and immunocytochemical staining. The scaffold facilitated the adhesion of corneal stromal cells, supporting cell proliferation, maintaining normal cytoskeletal organization, and promoting increased expression of genes associated with healthy corneal stromal keratocytes. These findings highlight the potential of a surface-modified PCL/SF blend (1:1) as a promising scaffolding material for corneal stromal regeneration. The developed scaffold not only demonstrated favourable biological interactions with corneal stromal cells but also exhibited characteristics aligned with the requirements for successful corneal tissue engineering. Further research and refinement of these constructs could lead to significant advancements in addressing the shortage of corneas for transplantation, ultimately improving the treatment outcomes for patients in need.

用于移植的人类角膜供体的短缺需要探索组织工程方法来开发角膜替代品。然而,这些替代品必须具备必要的强度、透明度和调节细胞行为的能力,才能用于患者。在本研究中,我们研究了氧等离子体表面修饰聚ε-己内酯(PCL)与丝素蛋白(SF)复合纳米纤维支架用于角膜基质再生的效果。在旋转芯轴上使用PCL和SF共混物制备电纺丝支架。该混合物的优化旨在复制人类角膜的结构和功能特性,重点关注纳米纤维的排列、力学特性以及与人类角膜基质角质细胞的体外细胞相容性。支架的表面修饰提高了透明度,增强了细胞间的相互作用。基于评价,我们选择了PCL与SF 1:1的复合纳米纤维支架进行更全面的分析。通过细胞粘附、增殖、细胞骨架组织、基因表达和免疫细胞化学染色来评估角化细胞对支架的生物学反应。支架促进角膜基质细胞的粘附,支持细胞增殖,维持正常的细胞骨架组织,并促进健康角膜基质角质细胞相关基因的表达增加。这些发现突出了表面改性PCL/SF共混物(1:1)作为角膜基质再生的有前途的支架材料的潜力。所开发的支架不仅与角膜基质细胞表现出良好的生物相互作用,而且表现出与成功的角膜组织工程要求相一致的特性。这些结构的进一步研究和改进可能会在解决角膜移植短缺方面取得重大进展,最终改善有需要的患者的治疗效果。
{"title":"Optimization and evaluation of oxygen-plasma-modified, aligned, poly (Є-caprolactone) and silk fibroin nanofibrous scaffold for corneal stromal regeneration","authors":"Promita Bhattacharjee ,&nbsp;Peter W. Madden ,&nbsp;Enzo Patriarca ,&nbsp;Mark Ahearne","doi":"10.1016/j.bbiosy.2023.100083","DOIUrl":"10.1016/j.bbiosy.2023.100083","url":null,"abstract":"<div><p>The shortage of human donor corneas for transplantation necessitates the exploration of tissue engineering approaches to develop corneal substitutes. However, these substitutes must possess the necessary strength, transparency, and ability to regulate cell behaviour before they can be used in patients. In this study, we investigated the effectiveness of an oxygen plasma surface-modified poly-ε-caprolactone (PCL) combined with silk fibroin (SF) nanofibrous scaffold for corneal stromal regeneration. To fabricate the electrospun scaffolds, PCL and SF blends were used on a rotating mandrel. The optimization of the blend aimed to replicate the structural and functional properties of the human cornea, focusing on nanofibre alignment, mechanical characteristics, and <em>in vitro</em> cytocompatibility with human corneal stromal keratocytes. Surface modification of the scaffold resulted in improved transparency and enhanced cell interaction. Based on the evaluation, a composite nanofibrous scaffold with a 1:1 blend of PCL and SF was selected for a more comprehensive analysis. The biological response of keratocytes to the scaffold was assessed through cellular adhesion, proliferation, cytoskeletal organization, gene expression, and immunocytochemical staining. The scaffold facilitated the adhesion of corneal stromal cells, supporting cell proliferation, maintaining normal cytoskeletal organization, and promoting increased expression of genes associated with healthy corneal stromal keratocytes. These findings highlight the potential of a surface-modified PCL/SF blend (1:1) as a promising scaffolding material for corneal stromal regeneration. The developed scaffold not only demonstrated favourable biological interactions with corneal stromal cells but also exhibited characteristics aligned with the requirements for successful corneal tissue engineering. Further research and refinement of these constructs could lead to significant advancements in addressing the shortage of corneas for transplantation, ultimately improving the treatment outcomes for patients in need.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/9c/cc/main.PMC10507194.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41158841","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mesenchymal stromal cell exosomes enhance dental pulp cell functions and promote pulp-dentin regeneration 间充质间质细胞外泌体增强牙髓细胞功能,促进牙髓-牙本质再生
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-09-01 DOI: 10.1016/j.bbiosy.2023.100078
Jiajun Shi , Kristeen Ye Wen Teo , Shipin Zhang , Ruenn Chai Lai , Vinicius Rosa , Huei Jinn Tong , Mandeep S. Duggal , Sai Kiang Lim , Wei Seong Toh

Mesenchymal stromal/stem cell (MSC) therapies are currently being explored for dental pulp regeneration. As the therapeutic effects of MSCs in tissue repair are mediated mainly through the release of extracellular vesicles (EVs) including exosomes, we investigated here the cellular processes and molecular mechanisms modulated by MSC exosomes in dental pulp regeneration. Using dental pulp cell (DPC) cultures, we demonstrated that MSC exosomes could increase DPC migration, proliferation, and odontogenic differentiation. The enhancement of these cellular processes was mediated through exosomal CD73-mediated adenosine receptor activation of AKT and ERK signaling. Consistent with these observations, MSC exosomes increased the expression of dentin matrix proteins and promoted the formation of dentin-like tissue and bridge-like structures in a rat pulp defect model. These effects were comparable to that of mineral trioxide aggregate (MTA) treatment. MSC exosomes also yielded recellularized pulp-dentin tissues in the root canal of endodontically-treated human premolars, following subcutaneous implantation in the mouse dorsum. Together, our findings suggest that MSC exosomes could exert a multi-faceted effect on DPC functions including migration, proliferation and odontogenic differentiation to promote dental pulp regeneration. This study provides the basis for development of MSC exosomes as a cell-free MSC therapeutic alternative for pulp-dentin regeneration.

间充质基质/干细胞(MSC)疗法目前正在探索用于牙髓再生。由于MSC在组织修复中的治疗作用主要通过包括外泌体在内的细胞外囊泡(EVs)的释放介导,我们在此研究了MSC外泌体在牙髓再生中调节的细胞过程和分子机制。使用牙髓细胞(DPC)培养,我们证明MSC外泌体可以增加DPC的迁移、增殖和牙源性分化。这些细胞过程的增强是通过外泌体CD73介导的AKT和ERK信号的腺苷受体激活介导的。与这些观察结果一致,在大鼠牙髓缺损模型中,MSC外泌体增加了牙本质基质蛋白的表达,并促进了牙本质样组织和桥状结构的形成。这些效果与矿物三氧化二聚集体(MTA)处理相当。MSC外泌体在小鼠背部皮下植入后,在根管治疗的人类前磨牙的根管中也产生了再细胞化的牙髓-牙本质组织。总之,我们的研究结果表明,MSC外泌体可以对DPC的功能发挥多方面的作用,包括迁移、增殖和牙源性分化,以促进牙髓再生。本研究为开发MSC外泌体作为牙髓-牙本质再生的无细胞MSC治疗替代品提供了基础。
{"title":"Mesenchymal stromal cell exosomes enhance dental pulp cell functions and promote pulp-dentin regeneration","authors":"Jiajun Shi ,&nbsp;Kristeen Ye Wen Teo ,&nbsp;Shipin Zhang ,&nbsp;Ruenn Chai Lai ,&nbsp;Vinicius Rosa ,&nbsp;Huei Jinn Tong ,&nbsp;Mandeep S. Duggal ,&nbsp;Sai Kiang Lim ,&nbsp;Wei Seong Toh","doi":"10.1016/j.bbiosy.2023.100078","DOIUrl":"10.1016/j.bbiosy.2023.100078","url":null,"abstract":"<div><p>Mesenchymal stromal/stem cell (MSC) therapies are currently being explored for dental pulp regeneration. As the therapeutic effects of MSCs in tissue repair are mediated mainly through the release of extracellular vesicles (EVs) including exosomes, we investigated here the cellular processes and molecular mechanisms modulated by MSC exosomes in dental pulp regeneration. Using dental pulp cell (DPC) cultures, we demonstrated that MSC exosomes could increase DPC migration, proliferation, and odontogenic differentiation. The enhancement of these cellular processes was mediated through exosomal CD73-mediated adenosine receptor activation of AKT and ERK signaling. Consistent with these observations, MSC exosomes increased the expression of dentin matrix proteins and promoted the formation of dentin-like tissue and bridge-like structures in a rat pulp defect model. These effects were comparable to that of mineral trioxide aggregate (MTA) treatment. MSC exosomes also yielded recellularized pulp-dentin tissues in the root canal of endodontically-treated human premolars, following subcutaneous implantation in the mouse dorsum. Together, our findings suggest that MSC exosomes could exert a multi-faceted effect on DPC functions including migration, proliferation and odontogenic differentiation to promote dental pulp regeneration. This study provides the basis for development of MSC exosomes as a cell-free MSC therapeutic alternative for pulp-dentin regeneration.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/33/ef/main.PMC10239699.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9592197","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Physicochemical cues are not potent regulators of human dermal fibroblast trans-differentiation 物理化学线索不是人类真皮成纤维细胞转分化的有效调节因子。
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-09-01 DOI: 10.1016/j.bbiosy.2023.100079
Christina N.M. Ryan , Eugenia Pugliese , Naledi Shologu , Diana Gaspar , Peadar Rooney , Md Nahidul Islam , Alan O'Riordan , Manus J. Biggs , Matthew D. Griffin , Dimitrios I. Zeugolis

Due to their inherent plasticity, dermal fibroblasts hold great promise in regenerative medicine. Although biological signals have been well-established as potent regulators of dermal fibroblast function, it is still unclear whether physiochemical cues can induce dermal fibroblast trans-differentiation. Herein, we evaluated the combined effect of surface topography, substrate rigidity, collagen type I coating and macromolecular crowding in human dermal fibroblast cultures. Our data indicate that tissue culture plastic and collagen type I coating increased cell proliferation and metabolic activity. None of the assessed in vitro microenvironment modulators affected cell viability. Anisotropic surface topography induced bidirectional cell morphology, especially on more rigid (1,000 kPa and 130 kPa) substrates. Macromolecular crowding increased various collagen types, but not fibronectin, deposition. Macromolecular crowding induced globular extracellular matrix deposition, independently of the properties of the substrate. At day 14 (longest time point assessed), macromolecular crowding downregulated tenascin C (in 9 out of the 14 groups), aggrecan (in 13 out of the 14 groups), osteonectin (in 13 out of the 14 groups), and collagen type I (in all groups). Overall, our data suggest that physicochemical cues (such surface topography, substrate rigidity, collagen coating and macromolecular crowding) are not as potent as biological signals in inducing dermal fibroblast trans-differentiation.

由于其固有的可塑性,真皮成纤维细胞在再生医学中具有很大的前景。尽管生物信号已被公认为真皮成纤维细胞功能的有效调节因子,但尚不清楚物理化学信号是否能诱导真皮成纤维纤维细胞的转分化。在此,我们评估了表面形貌、基质硬度、I型胶原涂层和大分子拥挤对人类真皮成纤维细胞培养的综合影响。我们的数据表明,组织培养塑料和I型胶原涂层增加了细胞增殖和代谢活性。所评估的体外微环境调节剂均未影响细胞活力。各向异性表面形貌诱导了双向细胞形态,尤其是在更刚性(1000kPa和130kPa)的基底上。大分子拥挤增加了各种胶原类型,但没有增加纤连蛋白的沉积。大分子拥挤诱导球状细胞外基质沉积,与基质的性质无关。在第14天(评估的最长时间点),大分子拥挤下调tenascin C(在14组中的9组中)、聚集蛋白聚糖(在14个组中的13组中),骨连蛋白(在14个中的13组)和I型胶原(在所有组中)。总体而言,我们的数据表明,物理化学线索(如表面形貌、基质硬度、胶原涂层和大分子拥挤)在诱导真皮成纤维细胞转分化方面不如生物信号有效。
{"title":"Physicochemical cues are not potent regulators of human dermal fibroblast trans-differentiation","authors":"Christina N.M. Ryan ,&nbsp;Eugenia Pugliese ,&nbsp;Naledi Shologu ,&nbsp;Diana Gaspar ,&nbsp;Peadar Rooney ,&nbsp;Md Nahidul Islam ,&nbsp;Alan O'Riordan ,&nbsp;Manus J. Biggs ,&nbsp;Matthew D. Griffin ,&nbsp;Dimitrios I. Zeugolis","doi":"10.1016/j.bbiosy.2023.100079","DOIUrl":"10.1016/j.bbiosy.2023.100079","url":null,"abstract":"<div><p>Due to their inherent plasticity, dermal fibroblasts hold great promise in regenerative medicine. Although biological signals have been well-established as potent regulators of dermal fibroblast function, it is still unclear whether physiochemical cues can induce dermal fibroblast trans-differentiation. Herein, we evaluated the combined effect of surface topography, substrate rigidity, collagen type I coating and macromolecular crowding in human dermal fibroblast cultures. Our data indicate that tissue culture plastic and collagen type I coating increased cell proliferation and metabolic activity. None of the assessed in vitro microenvironment modulators affected cell viability. Anisotropic surface topography induced bidirectional cell morphology, especially on more rigid (1,000 kPa and 130 kPa) substrates. Macromolecular crowding increased various collagen types, but not fibronectin, deposition. Macromolecular crowding induced globular extracellular matrix deposition, independently of the properties of the substrate. At day 14 (longest time point assessed), macromolecular crowding downregulated tenascin C (in 9 out of the 14 groups), aggrecan (in 13 out of the 14 groups), osteonectin (in 13 out of the 14 groups), and collagen type I (in all groups). Overall, our data suggest that physicochemical cues (such surface topography, substrate rigidity, collagen coating and macromolecular crowding) are not as potent as biological signals in inducing dermal fibroblast trans-differentiation.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10499661/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10289871","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Biomaterials are the key to unlock spheroid function and therapeutic potential 生物材料是开启球体功能和治疗潜力的关键。
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-09-01 DOI: 10.1016/j.bbiosy.2023.100080
David H. Ramos-Rodriguez , J. Kent Leach

Spheroids are three-dimensional cell aggregates that mimic fundamental aspects of the native tissue microenvironment better than single cells, making them a promising platform for the study of tissue development and therapeutics. Spheroids have been investigated for decades as models in cancer research, yet we have only just scratched the surface of their potential clinical utility in cell-based therapies. Like many cells, spheroids commonly exhibit a loss of key attributes upon implantation, motivating the need for strategies to regulate their function in situ. Biomaterials offer numerous opportunities to preserve spheroid function and guide spheroid behavior by tailoring the local microenvironment.

球体是三维细胞聚集体,比单细胞更好地模拟天然组织微环境的基本方面,使其成为研究组织发育和治疗的一个有前途的平台。几十年来,球状体一直被作为癌症研究的模型进行研究,但我们对其在基于细胞的治疗中的潜在临床应用还只是触及了表面。与许多细胞一样,球体在植入时通常表现出关键属性的丧失,这促使人们需要原位调节其功能的策略。生物材料通过调整局部微环境,为保持球体功能和指导球体行为提供了许多机会。
{"title":"Biomaterials are the key to unlock spheroid function and therapeutic potential","authors":"David H. Ramos-Rodriguez ,&nbsp;J. Kent Leach","doi":"10.1016/j.bbiosy.2023.100080","DOIUrl":"10.1016/j.bbiosy.2023.100080","url":null,"abstract":"<div><p>Spheroids are three-dimensional cell aggregates that mimic fundamental aspects of the native tissue microenvironment better than single cells, making them a promising platform for the study of tissue development and therapeutics. Spheroids have been investigated for decades as models in cancer research, yet we have only just scratched the surface of their potential clinical utility in cell-based therapies. Like many cells, spheroids commonly exhibit a loss of key attributes upon implantation, motivating the need for strategies to regulate their function <em>in situ</em>. Biomaterials offer numerous opportunities to preserve spheroid function and guide spheroid behavior by tailoring the local microenvironment.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10499638/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10290855","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Electrospun decellularized extracellular matrix scaffolds promote the regeneration of injured neurons 电纺丝脱细胞细胞外基质支架促进损伤神经元的再生
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-09-01 DOI: 10.1016/j.bbiosy.2023.100081
Lena Mungenast , Ronya Nieminen , Carine Gaiser , Ana Bela Faia-Torres , Jürgen Rühe , Laura Suter-Dick

Traumatic injury to the spinal cord (SCI) causes the transection of neurons, formation of a lesion cavity, and remodeling of the microenvironment by excessive extracellular matrix (ECM) deposition and scar formation leading to a regeneration-prohibiting environment. Electrospun fiber scaffolds have been shown to simulate the ECM and increase neural alignment and neurite outgrowth contributing to a growth-permissive matrix. In this work, electrospun ECM-like fibers providing biochemical and topological cues are implemented into a scaffold to represent an oriented biomaterial suitable for the alignment and migration of neural cells in order to improve spinal cord regeneration. The successfully decellularized spinal cord ECM (dECM), with no visible cell nuclei and dsDNA content < 50 ng/mg tissue, showed preserved ECM components, such as glycosaminoglycans and collagens. Serving as the biomaterial for 3D printer-assisted electrospinning, highly aligned and randomly distributed dECM fiber scaffolds (< 1 µm fiber diameter) were fabricated. The scaffolds were cytocompatible and supported the viability of a human neural cell line (SH-SY5Y) for 14 days. Cells were selectively differentiated into neurons, as confirmed by immunolabeling of specific cell markers (ChAT, Tubulin ß), and followed the orientation given by the dECM scaffolds. After generating a lesion site on the cell-scaffold model, cell migration was observed and compared to reference poly-ε-caprolactone fiber scaffolds. The aligned dECM fiber scaffold promoted the fastest and most efficient lesion closure, indicating superior cell guiding capabilities of dECM-based scaffolds. The strategy of combining decellularized tissues with controlled deposition of fibers to optimize biochemical and topographical cues opens the way for clinically relevant central nervous system scaffolding solutions.

脊髓创伤(SCI)导致神经元横断,形成病变腔,并通过过度的细胞外基质(ECM)沉积和瘢痕形成重塑微环境,从而导致再生禁止环境。电纺纤维支架已被证明可以模拟ECM,并增加神经排列和突起生长,从而形成生长允许基质。在这项工作中,将提供生物化学和拓扑线索的电纺ECM样纤维应用于支架中,以代表一种适用于神经细胞排列和迁移的定向生物材料,从而改善脊髓再生。成功脱细胞的脊髓ECM(dECM)没有可见的细胞核和dsDNA含量<;50ng/mg组织,显示保存的ECM成分,如糖胺聚糖和胶原。作为3D打印机辅助静电纺丝的生物材料,制备了高度排列和随机分布的dECM纤维支架(<;1µm纤维直径)。该支架具有细胞相容性,并支持人神经细胞系(SH-SY5Y)的生存能力达14天。细胞选择性分化为神经元,这通过特异性细胞标记物(ChAT,Tubulinß)的免疫标记得到证实,并遵循dECM支架给出的方向。在细胞支架模型上产生损伤部位后,观察细胞迁移,并与参考聚ε-己内酯纤维支架进行比较。排列的dECM纤维支架促进了最快、最有效的病变闭合,表明基于dECM的支架具有优越的细胞引导能力。将脱细胞组织与纤维的受控沉积相结合以优化生物化学和地形线索的策略为临床相关的中枢神经系统支架解决方案开辟了道路。
{"title":"Electrospun decellularized extracellular matrix scaffolds promote the regeneration of injured neurons","authors":"Lena Mungenast ,&nbsp;Ronya Nieminen ,&nbsp;Carine Gaiser ,&nbsp;Ana Bela Faia-Torres ,&nbsp;Jürgen Rühe ,&nbsp;Laura Suter-Dick","doi":"10.1016/j.bbiosy.2023.100081","DOIUrl":"10.1016/j.bbiosy.2023.100081","url":null,"abstract":"<div><p>Traumatic injury to the spinal cord (SCI) causes the transection of neurons, formation of a lesion cavity, and remodeling of the microenvironment by excessive extracellular matrix (ECM) deposition and scar formation leading to a regeneration-prohibiting environment. Electrospun fiber scaffolds have been shown to simulate the ECM and increase neural alignment and neurite outgrowth contributing to a growth-permissive matrix. In this work, electrospun ECM-like fibers providing biochemical and topological cues are implemented into a scaffold to represent an oriented biomaterial suitable for the alignment and migration of neural cells in order to improve spinal cord regeneration. The successfully decellularized spinal cord ECM (dECM), with no visible cell nuclei and dsDNA content &lt; 50 ng/mg tissue, showed preserved ECM components, such as glycosaminoglycans and collagens. Serving as the biomaterial for 3D printer-assisted electrospinning, highly aligned and randomly distributed dECM fiber scaffolds (&lt; 1 µm fiber diameter) were fabricated. The scaffolds were cytocompatible and supported the viability of a human neural cell line (SH-SY5Y) for 14 days. Cells were selectively differentiated into neurons, as confirmed by immunolabeling of specific cell markers (ChAT, Tubulin ß), and followed the orientation given by the dECM scaffolds. After generating a lesion site on the cell-scaffold model, cell migration was observed and compared to reference poly-ε-caprolactone fiber scaffolds. The aligned dECM fiber scaffold promoted the fastest and most efficient lesion closure, indicating superior cell guiding capabilities of dECM-based scaffolds. The strategy of combining decellularized tissues with controlled deposition of fibers to optimize biochemical and topographical cues opens the way for clinically relevant central nervous system scaffolding solutions.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/9e/8e/main.PMC10329103.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9811693","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
Disruptive 3D in vitro models for respiratory disease investigation: A state-of-the-art approach focused on SARS-CoV-2 infection 用于呼吸道疾病调查的破坏性3D体外模型:一种专注于SARS-CoV-2感染的最先进方法
Q3 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-09-01 DOI: 10.1016/j.bbiosy.2023.100082
Maria Luiza Seixas , Cynthia Silva Bartolomeo , Robertha Lemes , Tiago Nicoliche , Liria Hiromi Okuda , Leonardo Martins , Rodrigo Ureshino , Carla Maximo Prado , Tácia Tavares Aquinas Liguori , Gabriel Romero Liguori , Roberta Sessa Stilhano

COVID-19, along with most respiratory diseases in the medical field, demonstrates significant ability to take its toll on global population. There is a particular difficulty in studying these conditions, which stems especially from the short supply of in vitro models for detailed investigation, the specific therapeutic knowledge required for disease scrutinization and the occasional need of BSL-3 [Biosafety Level 3] laboratories for research. Based on this, the process of drug development is hampered to a great extent. In the scenario of COVID-19, this difficulty is even more substantial on account of the current undefinition regarding the exact role of the ACE2 [Angiotensin-converting enzyme 2] receptor upon SARS-CoV-2 kinetics in human cells and the great level of demand in the investigation process of ACE2, which usually requires the laborious and ethically complicated usage of transgenic animal models overexpressing the receptor. Moreover, the rapid progression of the aforementioned diseases, especially COVID-19, poses a crucial necessity for adequate therapeutic solutions emergence. In this context, the work herein presented introduces a groundbreaking set of 3D models, namely spheroids and MatriWell cell culture inserts, whose remarkable ability to mimic the in vivo environment makes them highly suitable for respiratory diseases investigation, particularly SARS-CoV-2 infection. Using MatriWells, we developed an innovative platform for COVID-19 research: a pulmonary air-liquid interface [ALI] associated with endothelial (HUVEC) cells. Infection studies revealed that pulmonary (BEAS-2B) cells in the ALI reached peak viral load at 24h and endothelial cells, at 48h, demonstrating lung viral replication and subsequent hematogenous dissemination, which provides us with a unique and realistic framework for studying COVID-19. Simultaneously, the spheroids were used to address the understudied ACE2 receptor, aiming at a pronounced process of COVID-19 investigation. ACE2 expression not only increased spheroid diameter by 20% (p<0.001) and volume by 60% (p≤0.0001) but also led to a remarkable 640-fold increase in intracellular viral load (p≤0.01). The previously mentioned finding supports ACE2 as a potential target for COVID-19 treatment. Lastly, we observed a higher viral load in the MatriWells compared to spheroids (150-fold, p<0.0001), suggesting the MatriWells as a more appropriate approach for COVID-19 investigation. By establishing an advanced method for respiratory tract conditions research, this work paves the way toward an efficacious process of drug development, contributing to a change in the course of respiratory diseases such as COVID-19.

新冠肺炎,以及医疗领域的大多数呼吸道疾病,显示出对全球人口造成重大损失的能力。研究这些条件特别困难,这尤其源于缺乏用于详细研究的体外模型、疾病检查所需的特定治疗知识,以及偶尔需要BSL-3[生物安全3级]实验室进行研究。基于此,药物开发过程在很大程度上受到阻碍。在新冠肺炎的情况下,由于目前关于ACE2(血管紧张素转化酶2)受体在人类细胞中的SARS-CoV-2动力学中的确切作用以及ACE2的研究过程中的巨大需求,这一困难更加严重,这通常需要费力且合乎伦理的使用过表达受体的转基因动物模型。此外,上述疾病,特别是新冠肺炎的迅速发展,对出现足够的治疗方案提出了至关重要的必要性。在此背景下,本文介绍了一组开创性的3D模型,即球体和MatriWell细胞培养插入物,其模拟体内环境的非凡能力使其非常适合呼吸系统疾病研究,特别是严重急性呼吸系统综合征冠状病毒2型感染。利用MatriWells,我们为新冠肺炎研究开发了一个创新平台:与内皮细胞(HUVEC)相关的肺气液界面[ALI]。感染研究表明,ALI中的肺细胞(BEAS-2B)在24小时达到病毒载量峰值,内皮细胞在48小时达到病毒负荷峰值,显示出肺病毒复制和随后的血行扩散,这为我们研究新冠肺炎提供了一个独特而现实的框架。同时,球体被用于解决研究不足的ACE2受体,旨在研究新冠肺炎的显著过程。ACE2的表达不仅使球体直径增加了20%(p<0.001),体积增加了60%(p≤0.0001),而且导致细胞内病毒载量显著增加了640倍(p≤0.01)。上述发现支持ACE2作为新冠肺炎治疗的潜在靶点。最后,我们观察到与球体相比,MatriWells中的病毒载量更高(150倍,p<0.0001),这表明MatriWelles是新冠肺炎调查的更合适的方法。通过建立一种先进的呼吸道疾病研究方法,这项工作为药物开发的有效过程铺平了道路,有助于改变新冠肺炎等呼吸道疾病的进程。
{"title":"Disruptive 3D in vitro models for respiratory disease investigation: A state-of-the-art approach focused on SARS-CoV-2 infection","authors":"Maria Luiza Seixas ,&nbsp;Cynthia Silva Bartolomeo ,&nbsp;Robertha Lemes ,&nbsp;Tiago Nicoliche ,&nbsp;Liria Hiromi Okuda ,&nbsp;Leonardo Martins ,&nbsp;Rodrigo Ureshino ,&nbsp;Carla Maximo Prado ,&nbsp;Tácia Tavares Aquinas Liguori ,&nbsp;Gabriel Romero Liguori ,&nbsp;Roberta Sessa Stilhano","doi":"10.1016/j.bbiosy.2023.100082","DOIUrl":"10.1016/j.bbiosy.2023.100082","url":null,"abstract":"<div><p>COVID-19, along with most respiratory diseases in the medical field, demonstrates significant ability to take its toll on global population. There is a particular difficulty in studying these conditions, which stems especially from the short supply of <em>in vitro</em> models for detailed investigation, the specific therapeutic knowledge required for disease scrutinization and the occasional need of BSL-3 [Biosafety Level 3] laboratories for research. Based on this, the process of drug development is hampered to a great extent. In the scenario of COVID-19, this difficulty is even more substantial on account of the current undefinition regarding the exact role of the ACE2 [Angiotensin-converting enzyme 2] receptor upon SARS-CoV-2 kinetics in human cells and the great level of demand in the investigation process of ACE2, which usually requires the laborious and ethically complicated usage of transgenic animal models overexpressing the receptor. Moreover, the rapid progression of the aforementioned diseases, especially COVID-19, poses a crucial necessity for adequate therapeutic solutions emergence. In this context, the work herein presented introduces a groundbreaking set of 3D models, namely spheroids and MatriWell cell culture inserts, whose remarkable ability to mimic the in vivo environment makes them highly suitable for respiratory diseases investigation, particularly SARS-CoV-2 infection. Using MatriWells, we developed an innovative platform for COVID-19 research: a pulmonary air-liquid interface [ALI] associated with endothelial (HUVEC) cells. Infection studies revealed that pulmonary (BEAS-2B) cells in the ALI reached peak viral load at 24h and endothelial cells, at 48h, demonstrating lung viral replication and subsequent hematogenous dissemination, which provides us with a unique and realistic framework for studying COVID-19. Simultaneously, the spheroids were used to address the understudied ACE2 receptor, aiming at a pronounced process of COVID-19 investigation. ACE2 expression not only increased spheroid diameter by 20% (p&lt;0.001) and volume by 60% (p≤0.0001) but also led to a remarkable 640-fold increase in intracellular viral load (p≤0.01). The previously mentioned finding supports ACE2 as a potential target for COVID-19 treatment. Lastly, we observed a higher viral load in the MatriWells compared to spheroids (150-fold, p&lt;0.0001), suggesting the MatriWells as a more appropriate approach for COVID-19 investigation. By establishing an advanced method for respiratory tract conditions research, this work paves the way toward an efficacious process of drug development, contributing to a change in the course of respiratory diseases such as COVID-19.</p></div>","PeriodicalId":72379,"journal":{"name":"Biomaterials and biosystems","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/e9/93/main.PMC10391659.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10308365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Biomaterials and biosystems
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: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