首页 > 最新文献

AMI: Acta Biomaterialia最新文献

英文 中文
Transepithelial Delivery of Insulin Conjugated with Phospholipid-Mimicking Polymers via Biomembrane Fusion-Mediated Transcellular Pathways 通过生物膜融合介导的跨细胞途径,胰岛素与磷脂模拟聚合物偶联经上皮传递
Pub Date : 2021-12-01 DOI: 10.2139/ssrn.3890358
Hiroaki Hatano, Fanlu Meng, Momoko Sakata, A. Matsumoto, K. Ishihara, Y. Miyahara, T. Goda
Epithelial barriers that seal cell gaps by forming tight junctions to prevent the free permeation of nutrients, electrolytes, and drugs, are essential for maintaining homeostasis in multicellular organisms. The development of nanocarriers that can permeate epithelial tissues without compromising barrier function is key for establishing a safe and efficient drug delivery system (DDS). Previously, we have demonstrated that a water-soluble phospholipid-mimicking random copolymer, poly(2-methacryloyloxyethyl phosphorylcholine30-random-n-butyl methacrylate70) (PMB30W), enters the cytoplasm of live cells by passive diffusion mechanisms, without damaging the cell membranes. The internalization mechanism was confirmed to be amphiphilicity-induced membrane fusion. In the present study, we demonstrated energy-independent permeation of PMB30W through the model epithelial barriers of Madin-Darby canine kidney (MDCK) cell monolayers in vitro. The polymer penetrated epithelial MDCK monolayers via transcellular pathways without breaching the barrier functions. This was confirmed by our unique assay that can monitor the leakage of the proton as the smallest indicator across the epithelial barriers. Moreover, energy-independent transepithelial permeation was achieved when insulin was chemically conjugated with the phospholipid-mimicking nanocarrier. The bioactivity of insulin as a growth factor was found to be maintained even after translocation. These fundamental findings may aid the establishment of transepithelial DDS with advanced drug efficiency and safety. STATEMENT OF SIGNIFICANCE: A nanocarrier that can freely permeate epithelial tissues without compromising barrier function is key for successful drug delivery system (DDS). Existing strategies mainly rely on paracellular transport associated with tight junction breakdown or transcellular transport via transporter recognition-mediated active uptake. These approaches raise concerns about efficiency and safety. In this study, we performed non-endocytic permeation of phospholipid-mimicking polymers through the model epithelial barriers in vitro. The polymer penetrated via transcytotic pathways without breaching the barriers of biomembrane and tight junction. Moreover, transepithelial permeation occurred when insulin was covalently attached to the nanocarrier. The bioactivity of insulin as a growth factor was maintained even after translocation. The biomimetic design of nanocarrier may realize safe and efficient transepithelial DDS.
上皮屏障通过形成紧密连接来封闭细胞间隙,以防止营养物质、电解质和药物的自由渗透,这对于维持多细胞生物的体内平衡至关重要。开发能够渗透上皮组织而不影响屏障功能的纳米载体是建立安全有效的给药系统(DDS)的关键。之前,我们已经证明了一种水溶性的模仿磷脂的无规则共聚物,聚(2-甲基丙烯酰氧乙基磷酸胆碱30-随机正丁基甲基丙烯酸酯70)(PMB30W),通过被动扩散机制进入活细胞的细胞质,而不破坏细胞膜。内化机制证实为两亲性诱导的膜融合。在本研究中,我们在体外证明PMB30W通过Madin-Darby犬肾(MDCK)细胞单层模型上皮屏障的能量非依赖性渗透。聚合物通过跨细胞途径穿透上皮MDCK单层而不破坏屏障功能。我们独特的检测证实了这一点,该检测可以监测质子泄漏,作为跨越上皮屏障的最小指标。此外,当胰岛素与模拟磷脂的纳米载体化学偶联时,实现了能量独立的经上皮渗透。胰岛素作为一种生长因子的生物活性被发现即使在易位后也能保持。这些基本发现可能有助于建立具有先进药物效率和安全性的经上皮性DDS。意义声明:一种可以自由渗透上皮组织而不影响屏障功能的纳米载体是成功的药物递送系统(DDS)的关键。现有的策略主要依赖于与紧密连接破坏相关的细胞旁转运或通过转运体识别介导的主动摄取的跨细胞转运。这些方法引起了人们对效率和安全性的担忧。在这项研究中,我们在体外通过模型上皮屏障进行了磷脂模拟聚合物的非内吞渗透。聚合物在不破坏生物膜和紧密连接屏障的情况下,通过胞吞途径渗透。此外,当胰岛素与纳米载体共价结合时,会发生经上皮渗透。即使在易位后,胰岛素作为生长因子的生物活性仍保持不变。纳米载体的仿生设计可实现安全高效的经上皮DDS。
{"title":"Transepithelial Delivery of Insulin Conjugated with Phospholipid-Mimicking Polymers via Biomembrane Fusion-Mediated Transcellular Pathways","authors":"Hiroaki Hatano, Fanlu Meng, Momoko Sakata, A. Matsumoto, K. Ishihara, Y. Miyahara, T. Goda","doi":"10.2139/ssrn.3890358","DOIUrl":"https://doi.org/10.2139/ssrn.3890358","url":null,"abstract":"Epithelial barriers that seal cell gaps by forming tight junctions to prevent the free permeation of nutrients, electrolytes, and drugs, are essential for maintaining homeostasis in multicellular organisms. The development of nanocarriers that can permeate epithelial tissues without compromising barrier function is key for establishing a safe and efficient drug delivery system (DDS). Previously, we have demonstrated that a water-soluble phospholipid-mimicking random copolymer, poly(2-methacryloyloxyethyl phosphorylcholine30-random-n-butyl methacrylate70) (PMB30W), enters the cytoplasm of live cells by passive diffusion mechanisms, without damaging the cell membranes. The internalization mechanism was confirmed to be amphiphilicity-induced membrane fusion. In the present study, we demonstrated energy-independent permeation of PMB30W through the model epithelial barriers of Madin-Darby canine kidney (MDCK) cell monolayers in vitro. The polymer penetrated epithelial MDCK monolayers via transcellular pathways without breaching the barrier functions. This was confirmed by our unique assay that can monitor the leakage of the proton as the smallest indicator across the epithelial barriers. Moreover, energy-independent transepithelial permeation was achieved when insulin was chemically conjugated with the phospholipid-mimicking nanocarrier. The bioactivity of insulin as a growth factor was found to be maintained even after translocation. These fundamental findings may aid the establishment of transepithelial DDS with advanced drug efficiency and safety. STATEMENT OF SIGNIFICANCE: A nanocarrier that can freely permeate epithelial tissues without compromising barrier function is key for successful drug delivery system (DDS). Existing strategies mainly rely on paracellular transport associated with tight junction breakdown or transcellular transport via transporter recognition-mediated active uptake. These approaches raise concerns about efficiency and safety. In this study, we performed non-endocytic permeation of phospholipid-mimicking polymers through the model epithelial barriers in vitro. The polymer penetrated via transcytotic pathways without breaching the barriers of biomembrane and tight junction. Moreover, transepithelial permeation occurred when insulin was covalently attached to the nanocarrier. The bioactivity of insulin as a growth factor was maintained even after translocation. The biomimetic design of nanocarrier may realize safe and efficient transepithelial DDS.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"125591747","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Magnetic Nanocomposite Hydrogel with Tunable Stiffness for Probing Cellular Responses to Matrix Stiffening 具有可调刚度的磁性纳米复合水凝胶用于探测细胞对基质硬化的反应
Pub Date : 2021-11-01 DOI: 10.2139/ssrn.3897769
Tianhao Yan, D. Rao, Ye Chen, Yu Wang, Qingchuan Zhang, Shangquan Wu
As cells have the capacity to respond to their mechanical environment, cellular biological behaviors can be regulated by the stiffness of extracellular matrix. Moreover, biological processes are dynamic and accompanied by matrix stiffening. Herein, we developed a stiffening cell culture platform based on polyacrylamide-Fe3O4 magnetic nanocomposite hydrogel with tunable stiffness under the application of magnetic field. This platform provided a wide range of tunable stiffness (∼0.3-20 kPa) covering most of human tissue elasticity with a high biocompatibility. Overall, the increased magnetic interactions between magnetic nanoparticles reduced the pore size of the hydrogel and enhanced the hydrogel stiffness, thereby facilitating the adhesion and spreading of stem cells, which was attributed to the F-actin assembly and vinculin recruitment. Such stiffening cell culture platform provides dynamic mechanical environments for probing the cellular response to matrix stiffening, and benefits studies of dynamic biological processes. STATEMENT OF SIGNIFICANCE: : Cellular biological behaviors can be regulated by the stiffness of extracellular matrix. Moreover, biological processes are dynamic and accompanied by matrix stiffening. Herein, we developed a stiffening cell culture platform based on polyacrylamide/Fe3O4 magnetic nanocomposite hydrogels with a wide tunable range of stiffness under the application of magnetic field, without adversely affecting cellular behaviors. Such matrix stiffening caused by enhanced magnetic interaction between magnetic nanoparticles under the application of the magnetic field could induce the morphological variations of stem cells cultured on the hydrogels. Overall, our stiffening cell culture platform can be used not only to probe the cellular response to matrix stiffening but also to benefit various biomedical studies.
由于细胞具有对其机械环境作出反应的能力,细胞的生物学行为可以通过细胞外基质的刚度来调节。此外,生物过程是动态的,并伴有基质硬化。为此,我们开发了一种基于聚丙烯酰胺- fe3o4磁性纳米复合水凝胶的增强细胞培养平台,该平台在磁场作用下具有可调的刚度。该平台提供了广泛的可调刚度(~ 0.3-20 kPa),覆盖了大部分人体组织弹性,具有高生物相容性。总的来说,磁性纳米颗粒之间增加的磁性相互作用减小了水凝胶的孔径,增强了水凝胶的硬度,从而促进了干细胞的粘附和扩散,这归因于f -肌动蛋白的组装和血管蛋白的募集。这种强化细胞培养平台为探索细胞对基质强化的反应提供了动态力学环境,有利于动态生物过程的研究。重要意义:细胞外基质的刚度可以调节细胞的生物学行为。此外,生物过程是动态的,并伴有基质硬化。在此,我们开发了一种基于聚丙烯酰胺/Fe3O4磁性纳米复合水凝胶的硬化细胞培养平台,该平台在磁场作用下具有宽范围可调的刚度,而不会对细胞行为产生不利影响。在磁场作用下,磁性纳米颗粒之间的磁相互作用增强,导致基质硬化,从而诱导水凝胶上培养的干细胞发生形态变化。总的来说,我们的硬化细胞培养平台不仅可以用来探测细胞对基质硬化的反应,还可以用于各种生物医学研究。
{"title":"Magnetic Nanocomposite Hydrogel with Tunable Stiffness for Probing Cellular Responses to Matrix Stiffening","authors":"Tianhao Yan, D. Rao, Ye Chen, Yu Wang, Qingchuan Zhang, Shangquan Wu","doi":"10.2139/ssrn.3897769","DOIUrl":"https://doi.org/10.2139/ssrn.3897769","url":null,"abstract":"As cells have the capacity to respond to their mechanical environment, cellular biological behaviors can be regulated by the stiffness of extracellular matrix. Moreover, biological processes are dynamic and accompanied by matrix stiffening. Herein, we developed a stiffening cell culture platform based on polyacrylamide-Fe3O4 magnetic nanocomposite hydrogel with tunable stiffness under the application of magnetic field. This platform provided a wide range of tunable stiffness (∼0.3-20 kPa) covering most of human tissue elasticity with a high biocompatibility. Overall, the increased magnetic interactions between magnetic nanoparticles reduced the pore size of the hydrogel and enhanced the hydrogel stiffness, thereby facilitating the adhesion and spreading of stem cells, which was attributed to the F-actin assembly and vinculin recruitment. Such stiffening cell culture platform provides dynamic mechanical environments for probing the cellular response to matrix stiffening, and benefits studies of dynamic biological processes. STATEMENT OF SIGNIFICANCE: : Cellular biological behaviors can be regulated by the stiffness of extracellular matrix. Moreover, biological processes are dynamic and accompanied by matrix stiffening. Herein, we developed a stiffening cell culture platform based on polyacrylamide/Fe3O4 magnetic nanocomposite hydrogels with a wide tunable range of stiffness under the application of magnetic field, without adversely affecting cellular behaviors. Such matrix stiffening caused by enhanced magnetic interaction between magnetic nanoparticles under the application of the magnetic field could induce the morphological variations of stem cells cultured on the hydrogels. Overall, our stiffening cell culture platform can be used not only to probe the cellular response to matrix stiffening but also to benefit various biomedical studies.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"115465887","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 14
Biomimetic Matrix for the Study of Neuroblastoma Cells: A Promising Combination of Stiffness and Retinoic Acid 用于神经母细胞瘤细胞研究的仿生基质:刚度和维甲酸的有前途的组合
Pub Date : 2021-08-01 DOI: 10.2139/ssrn.3849028
B. Labat, N. Buchbinder, S. Morin-Grognet, G. Ladam, Hassan Atmani, J. Vannier
Neuroblastoma is the third most common pediatric cancer composed of malignant immature cells that are usually treated pharmacologically by all trans-retinoic acid (ATRA) but sometimes, they can spontaneously differentiate into benign forms. In that context, biomimetic cell culture models are warranted tools as they can recapitulate many of the biochemical and biophysical cues of normal or pathological microenvironments. Inspired by that challenge, we developed a neuroblastoma culture system based on biomimetic LbL films of physiological biochemical composition and mechanical properties. For that, we used chondroitin sulfate A (CSA) and poly-L-lysine (PLL) that were assembled and mechanically tuned by crosslinking with genipin (GnP), a natural biocompatible crosslinker, in a relevant range of stiffness (30-160 kPa). We then assessed the adhesion, survival, motility, and differentiation of LAN-1 neuroblastoma cells. Remarkably, increasing the stiffness of the LbL films induced neuritogenesis that was strengthened by the combination with ATRA. These results highlight the crucial role of the mechanical cues of the neuroblastoma microenvironment since it can dramatically modulate the effect of pharmacologic drugs. In conclusion, our biomimetic platform offers a promising tool to help fundamental understanding and pharmacological screening of neuroblastoma differentiation and may assist the design of translational biomaterials to support neuronal regeneration. Statement of significance: Neuroblastoma is one of the most common pediatric tumor commonly treated by the administration of all-trans-retinoic acid (ATRA). Unfortunately, advanced neuroblastoma often develop ATRA resistance. Accordingly, in the field of pharmacological investigations on neuroblastoma, there is a tremendous need of physiologically relevant cell culture systems that can mimic normal or pathological extracellular matrices. In that context, we developed a promising matrix-like cell culture model that provides new insights on the crucial role of mechanical properties of the microenvironment upon the success of ATRA treatment on the neuroblastoma maturation. We were able to control adhesion, survival, motility, and differentiation of neuroblastoma cells. More broadly, we believe that our system will help the design of in vitro pharmacological screening strategy.
神经母细胞瘤是第三种最常见的儿科癌症,由恶性未成熟细胞组成,通常用全反式维甲酸(ATRA)治疗,但有时它们可以自发地分化为良性形式。在这种情况下,仿生细胞培养模型是可靠的工具,因为它们可以概括正常或病理微环境的许多生化和生物物理线索。受到这一挑战的启发,我们开发了一种基于生理生化组成和机械性能的仿生LbL膜的神经母细胞瘤培养系统。为此,我们使用硫酸软骨素A (CSA)和聚l -赖氨酸(PLL),它们通过与天然生物相容性交联剂genipin (GnP)在相关刚度范围(30-160 kPa)内交联进行组装和机械调谐。然后我们评估了LAN-1神经母细胞瘤细胞的粘附性、存活率、运动性和分化。值得注意的是,增加LbL膜的硬度诱导神经新生,与ATRA联合使用可加强神经新生。这些结果强调了神经母细胞瘤微环境的机械线索的关键作用,因为它可以显着调节药物的作用。总之,我们的仿生平台提供了一个很有前途的工具,有助于基本理解和神经母细胞瘤分化的药理学筛选,并可能有助于设计支持神经元再生的翻译生物材料。意义声明:神经母细胞瘤是最常见的儿科肿瘤之一,通常采用全反式维甲酸(ATRA)治疗。不幸的是,晚期神经母细胞瘤经常产生ATRA耐药性。因此,在神经母细胞瘤的药理学研究领域,迫切需要能够模拟正常或病理细胞外基质的生理相关细胞培养系统。在此背景下,我们开发了一种很有前景的基质样细胞培养模型,该模型为微环境的机械特性在ATRA治疗神经母细胞瘤成熟过程中成功发挥的关键作用提供了新的见解。我们能够控制成神经细胞瘤细胞的粘附、存活、运动和分化。更广泛地说,我们相信我们的系统将有助于体外药理学筛选策略的设计。
{"title":"Biomimetic Matrix for the Study of Neuroblastoma Cells: A Promising Combination of Stiffness and Retinoic Acid","authors":"B. Labat, N. Buchbinder, S. Morin-Grognet, G. Ladam, Hassan Atmani, J. Vannier","doi":"10.2139/ssrn.3849028","DOIUrl":"https://doi.org/10.2139/ssrn.3849028","url":null,"abstract":"Neuroblastoma is the third most common pediatric cancer composed of malignant immature cells that are usually treated pharmacologically by all trans-retinoic acid (ATRA) but sometimes, they can spontaneously differentiate into benign forms. In that context, biomimetic cell culture models are warranted tools as they can recapitulate many of the biochemical and biophysical cues of normal or pathological microenvironments. Inspired by that challenge, we developed a neuroblastoma culture system based on biomimetic LbL films of physiological biochemical composition and mechanical properties. For that, we used chondroitin sulfate A (CSA) and poly-L-lysine (PLL) that were assembled and mechanically tuned by crosslinking with genipin (GnP), a natural biocompatible crosslinker, in a relevant range of stiffness (30-160 kPa). We then assessed the adhesion, survival, motility, and differentiation of LAN-1 neuroblastoma cells. Remarkably, increasing the stiffness of the LbL films induced neuritogenesis that was strengthened by the combination with ATRA. These results highlight the crucial role of the mechanical cues of the neuroblastoma microenvironment since it can dramatically modulate the effect of pharmacologic drugs. In conclusion, our biomimetic platform offers a promising tool to help fundamental understanding and pharmacological screening of neuroblastoma differentiation and may assist the design of translational biomaterials to support neuronal regeneration. Statement of significance: Neuroblastoma is one of the most common pediatric tumor commonly treated by the administration of all-trans-retinoic acid (ATRA). Unfortunately, advanced neuroblastoma often develop ATRA resistance. Accordingly, in the field of pharmacological investigations on neuroblastoma, there is a tremendous need of physiologically relevant cell culture systems that can mimic normal or pathological extracellular matrices. In that context, we developed a promising matrix-like cell culture model that provides new insights on the crucial role of mechanical properties of the microenvironment upon the success of ATRA treatment on the neuroblastoma maturation. We were able to control adhesion, survival, motility, and differentiation of neuroblastoma cells. More broadly, we believe that our system will help the design of in vitro pharmacological screening strategy.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"126472054","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
Multi-Functional Silica-Based Mesoporous Materials as Co-Delivery Systems for Biologically Active Ions and Therapeutic Biomolecules 多功能硅基介孔材料作为生物活性离子和治疗性生物分子的共递送系统
Pub Date : 2021-01-21 DOI: 10.2139/ssrn.3770987
Hui Zhu, K. Zheng, A. Boccaccini
Mesoporous silica-based materials, especially mesoporous bioactive glasses (MBGs), are widely used in biomedical applications including tissue engineering and drug delivery, not only because of their excellent bioactivity and biocompatibility but also due to their tunable composition and potential use as drug delivery carriers owing to their controllable nanoporous structure. Numerous researches have reported that MBGs can be doped with various therapeutic ions (strontium, copper, magnesium, zinc, lithium, silver, etc.) and loaded with specific biomolecules (e.g., therapeutic drugs, antibiotics, growth factors) achieving controllable loading and release kinetics. Therefore, co-delivery of ions and biomolecules using a single MBG carrier is highly interesting as this approach provides synergistic effects toward improved therapeutic outcomes in comparison to the strategy of sole drug or ion delivery. In this review, we discuss the state-of-the-art in the field of mesoporous silica-based materials used for co-delivery of ions and therapeutic drugs with osteogenesis/cementogenesis, angiogenesis, antibacterial and anticancer properties. The analysis of the literature reveals that specially designed mesoporous nanocarriers could controllably release multiple ions and drugs at therapeutically safe and relevant levels, achieving the desired biological effects (in vivo, in vitro) for specific biomedical applications. It is expected that this review on the ion/drug co-delivery concept using MBG carriers will shed light on the advantages of such co-delivery systems for clinical use. Areas for future research directions are identified and discussed.
介孔硅基材料,特别是介孔生物活性玻璃(MBGs),不仅因其优异的生物活性和生物相容性,而且由于其可调节的纳米孔结构和潜在的药物递送载体,在生物医学领域广泛应用于组织工程和药物递送。大量研究报道,MBGs可以掺杂各种治疗离子(锶、铜、镁、锌、锂、银等),并装载特定的生物分子(如治疗药物、抗生素、生长因子),实现可控的装载和释放动力学。因此,使用单一MBG载体的离子和生物分子的共同递送是非常有趣的,因为与单一药物或离子递送策略相比,这种方法提供了改善治疗结果的协同效应。在这篇综述中,我们讨论了中孔硅基材料在离子和治疗药物共递送领域的最新进展,这些材料具有成骨/骨水泥生成、血管生成、抗菌和抗癌等特性。通过对文献的分析发现,经过特殊设计的介孔纳米载体能够在治疗安全性和相关水平上可控地释放多种离子和药物,从而达到特定生物医学应用所需的生物效应(体内和体外)。这篇关于使用MBG载体的离子/药物共给药概念的综述将揭示这种共给药系统在临床应用中的优势。确定并讨论了今后的研究方向。
{"title":"Multi-Functional Silica-Based Mesoporous Materials as Co-Delivery Systems for Biologically Active Ions and Therapeutic Biomolecules","authors":"Hui Zhu, K. Zheng, A. Boccaccini","doi":"10.2139/ssrn.3770987","DOIUrl":"https://doi.org/10.2139/ssrn.3770987","url":null,"abstract":"Mesoporous silica-based materials, especially mesoporous bioactive glasses (MBGs), are widely used in biomedical applications including tissue engineering and drug delivery, not only because of their excellent bioactivity and biocompatibility but also due to their tunable composition and potential use as drug delivery carriers owing to their controllable nanoporous structure. Numerous researches have reported that MBGs can be doped with various therapeutic ions (strontium, copper, magnesium, zinc, lithium, silver, etc.) and loaded with specific biomolecules (e.g., therapeutic drugs, antibiotics, growth factors) achieving controllable loading and release kinetics. Therefore, co-delivery of ions and biomolecules using a single MBG carrier is highly interesting as this approach provides synergistic effects toward improved therapeutic outcomes in comparison to the strategy of sole drug or ion delivery. In this review, we discuss the state-of-the-art in the field of mesoporous silica-based materials used for co-delivery of ions and therapeutic drugs with osteogenesis/cementogenesis, angiogenesis, antibacterial and anticancer properties. The analysis of the literature reveals that specially designed mesoporous nanocarriers could controllably release multiple ions and drugs at therapeutically safe and relevant levels, achieving the desired biological effects (in vivo, in vitro) for specific biomedical applications. It is expected that this review on the ion/drug co-delivery concept using MBG carriers will shed light on the advantages of such co-delivery systems for clinical use. Areas for future research directions are identified and discussed.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"125386439","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
Targeted Brain Delivery of RVG29-Modified Rifampicin-Loaded Nanoparticles for Treatment and Diagnosis of Alzheimer Disease 靶向脑递送rvg29修饰的利福平纳米颗粒治疗和诊断阿尔茨海默病
Pub Date : 2021-01-07 DOI: 10.2139/ssrn.3762216
Ruiyi Zhou, Lihong Zhu, Zhaohao Zeng, Rixin Luo, Jiawei Zhang, Li Deng, R. Guo, Lei Zhang, Qunying Zhang, Wei Bi
Alzheimer's disease (AD) is a neurodegenerative disease related to age. The main pathological features of AD are β-amyloid protein (Aβ) deposition and tau protein hyperphosphorylation. Currently, there are not an effective drug for the etiological treatment of AD. Rifampicin (RIF) is a semi-synthetic broad-spectrum antibiotic with anti-β-amyloid deposition, anti-inflammatory, anti-apoptosis and neuroprotective effects, but its application in AD treatment has been limited by its strong hydrophobicity, high toxicity, short half-life, low bioavailability and blood-brain barrier (BBB) hindrance. We designed a novel brain-targeted and MRI-characteristic nanomedicine through loading rabies virus protein 29 (RVG29), rifampicin and Gd on poly(L-lactide) nanoparticles (RIF@PLA-PEG-Gd/Mal-RVG29). The cytotoxicity assay demonstrated that RIF@PLA-PEG-Gd/Mal-RVG29 had good biocompatibility and security. The results of cellular uptake and fluorescence in vivo imaging showed that PLA-PEG-Gd/Mal-RVG29 could deliver rifampicin into brain by enhancing cellular uptake and brain targeting performance, so that improving the bioavailability of rifampicin. In vivo study, compared with rifampicin, RIF@PLA-PEG-Gd/Mal-RVG29 improved the spatial learning and memory capability of APP/PS1 mice in the Morris water maze. Immunofluorescence, TEM, immunoblotting and HE test showed that RIF@PLA-PEG-Gd/Mal-RVG29 could reduce Aβ deposition in hippocampal and cortex of APP/PS1 mice, improve the damage of synaptic ultrastructure, increase the expression level of PSD95 and SYP, as well as reduce the necrosis of neurons. These findings suggest that RIF@PLA-PEG-Gd/Mal-RVG29 may be an effective strategy for the treatment and diagnosis of AD.
阿尔茨海默病(AD)是一种与年龄有关的神经退行性疾病。AD的主要病理特征是β-淀粉样蛋白(Aβ)沉积和tau蛋白过度磷酸化。目前,还没有一种有效的药物对阿尔茨海默病进行病因治疗。利福平(Rifampicin, RIF)是一种具有抗β-淀粉样蛋白沉积、抗炎、抗细胞凋亡和神经保护作用的半合成广谱抗生素,但其疏水性强、毒性高、半衰期短、生物利用度低、血脑屏障(BBB)阻碍等缺点限制了其在AD治疗中的应用。我们通过将狂犬病毒蛋白29 (RVG29)、利福平和Gd装载在聚(l-丙交酯)纳米颗粒(RIF@PLA-PEG-Gd/Mal-RVG29)上,设计了一种新的脑靶向和mri特征纳米药物。细胞毒性实验表明RIF@PLA-PEG-Gd/Mal-RVG29具有良好的生物相容性和安全性。细胞摄取和体内荧光成像结果显示,PLA-PEG-Gd/Mal-RVG29可通过增强细胞摄取和脑靶向性能将利福平输送到脑内,从而提高利福平的生物利用度。在体内研究中,RIF@PLA-PEG-Gd/Mal-RVG29与利福平相比,可提高APP/PS1小鼠Morris水迷宫的空间学习记忆能力。免疫荧光、透射电镜、免疫印迹和HE检测显示RIF@PLA-PEG-Gd/Mal-RVG29可减少APP/PS1小鼠海马和皮层Aβ沉积,改善突触超微结构损伤,提高PSD95和SYP表达水平,减轻神经元坏死。这些发现提示RIF@PLA-PEG-Gd/Mal-RVG29可能是治疗和诊断AD的有效策略。
{"title":"Targeted Brain Delivery of RVG29-Modified Rifampicin-Loaded Nanoparticles for Treatment and Diagnosis of Alzheimer Disease","authors":"Ruiyi Zhou, Lihong Zhu, Zhaohao Zeng, Rixin Luo, Jiawei Zhang, Li Deng, R. Guo, Lei Zhang, Qunying Zhang, Wei Bi","doi":"10.2139/ssrn.3762216","DOIUrl":"https://doi.org/10.2139/ssrn.3762216","url":null,"abstract":"Alzheimer's disease (AD) is a neurodegenerative disease related to age. The main pathological features of AD are β-amyloid protein (Aβ) deposition and tau protein hyperphosphorylation. Currently, there are not an effective drug for the etiological treatment of AD. Rifampicin (RIF) is a semi-synthetic broad-spectrum antibiotic with anti-β-amyloid deposition, anti-inflammatory, anti-apoptosis and neuroprotective effects, but its application in AD treatment has been limited by its strong hydrophobicity, high toxicity, short half-life, low bioavailability and blood-brain barrier (BBB) hindrance. We designed a novel brain-targeted and MRI-characteristic nanomedicine through loading rabies virus protein 29 (RVG29), rifampicin and Gd on poly(L-lactide) nanoparticles (RIF@PLA-PEG-Gd/Mal-RVG29). The cytotoxicity assay demonstrated that RIF@PLA-PEG-Gd/Mal-RVG29 had good biocompatibility and security. The results of cellular uptake and fluorescence in vivo imaging showed that PLA-PEG-Gd/Mal-RVG29 could deliver rifampicin into brain by enhancing cellular uptake and brain targeting performance, so that improving the bioavailability of rifampicin. In vivo study, compared with rifampicin, RIF@PLA-PEG-Gd/Mal-RVG29 improved the spatial learning and memory capability of APP/PS1 mice in the Morris water maze. Immunofluorescence, TEM, immunoblotting and HE test showed that RIF@PLA-PEG-Gd/Mal-RVG29 could reduce Aβ deposition in hippocampal and cortex of APP/PS1 mice, improve the damage of synaptic ultrastructure, increase the expression level of PSD95 and SYP, as well as reduce the necrosis of neurons. These findings suggest that RIF@PLA-PEG-Gd/Mal-RVG29 may be an effective strategy for the treatment and diagnosis of AD.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"131042740","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hydrogels for Large-Scale Expansion of Stem Cells 用于干细胞大规模扩增的水凝胶
Pub Date : 2021-01-07 DOI: 10.2139/ssrn.3762193
Sheng Yin, Yi Cao
Stem cells demonstrate considerable promise for various preclinical and clinical applications, including drug screening, disease treatments, and regenerative medicine. Producing high-quality and large amounts of stem cells is in demand for these applications. Despite challenges, as hydrogel-based cell culture technology has developed, tremendous progress has been made in stem cell expansion and directed differentiation. Hydrogels are soft materials with abundant water. Many hydrogel properties, including biodegradability, mechanical strength, and porosity, have been shown to play essential roles in regulating stem cell proliferation and differentiation. The biochemical and physical properties of hydrogels can be specifically tailored to mimic the native microenvironment that various stem cells reside in vivo. A few hydrogel-based systems have been developed for successful stem cell cultures and expansion in vitro. In this review, we summarize various types of hydrogels that have been designed to effectively enhance the proliferation of hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs), respectively. According to each stem cell type's preference, we also discuss strategies for fabricating hydrogels with biochemical and mechanical cues and other characteristics representing microenvironments of stem cells in vivo. STATEMENT OF SIGNIFICANCE: In this review article we summary current progress on the construction of hydrogel systems for the culture and expansion of various stem cells, including hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs). The Significance includes: (1) Provide detailed discussion on the stem cell niches that should be considered for stem cell in vitro expansion. (2) Summarize various strategies to construct hydrogels that can largely recapture the microenvironment of native stem cells. (3) Suggest a few future directions that can be implemented to improve current in vitro stem cell expansion systems.
干细胞在各种临床前和临床应用方面表现出相当大的前景,包括药物筛选、疾病治疗和再生医学。生产高质量和大量的干细胞是这些应用的需求。尽管存在诸多挑战,但随着水凝胶细胞培养技术的发展,干细胞扩增和定向分化取得了巨大进展。水凝胶是一种含有丰富水分的柔软材料。水凝胶的许多特性,包括生物降解性、机械强度和孔隙度,在调节干细胞增殖和分化中发挥着重要作用。水凝胶的生化和物理特性可以专门用于模拟各种干细胞在体内生存的原生微环境。一些基于水凝胶的系统已经开发成功的干细胞培养和体外扩增。在这篇综述中,我们分别总结了各种类型的水凝胶,它们被设计用来有效地促进造血干细胞(hsc)、间充质干细胞/基质细胞(MSCs)和多能干细胞(PSCs)的增殖。根据每种干细胞类型的偏好,我们还讨论了利用生物化学和机械线索以及代表干细胞体内微环境的其他特征制造水凝胶的策略。在这篇综述文章中,我们总结了目前用于培养和扩增各种干细胞的水凝胶体系的建设进展,包括造血干细胞(hsc),间充质干细胞/基质细胞(MSCs)和多能干细胞(PSCs)。其意义包括:(1)对干细胞体外扩增应考虑的干细胞壁龛进行了详细的讨论。(2)总结了构建水凝胶的各种策略,这些策略可以在很大程度上重现天然干细胞的微环境。(3)提出未来可以实施的几个方向,以改进现有的体外干细胞扩增系统。
{"title":"Hydrogels for Large-Scale Expansion of Stem Cells","authors":"Sheng Yin, Yi Cao","doi":"10.2139/ssrn.3762193","DOIUrl":"https://doi.org/10.2139/ssrn.3762193","url":null,"abstract":"Stem cells demonstrate considerable promise for various preclinical and clinical applications, including drug screening, disease treatments, and regenerative medicine. Producing high-quality and large amounts of stem cells is in demand for these applications. Despite challenges, as hydrogel-based cell culture technology has developed, tremendous progress has been made in stem cell expansion and directed differentiation. Hydrogels are soft materials with abundant water. Many hydrogel properties, including biodegradability, mechanical strength, and porosity, have been shown to play essential roles in regulating stem cell proliferation and differentiation. The biochemical and physical properties of hydrogels can be specifically tailored to mimic the native microenvironment that various stem cells reside in vivo. A few hydrogel-based systems have been developed for successful stem cell cultures and expansion in vitro. In this review, we summarize various types of hydrogels that have been designed to effectively enhance the proliferation of hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs), respectively. According to each stem cell type's preference, we also discuss strategies for fabricating hydrogels with biochemical and mechanical cues and other characteristics representing microenvironments of stem cells in vivo. STATEMENT OF SIGNIFICANCE: In this review article we summary current progress on the construction of hydrogel systems for the culture and expansion of various stem cells, including hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs). The Significance includes: (1) Provide detailed discussion on the stem cell niches that should be considered for stem cell in vitro expansion. (2) Summarize various strategies to construct hydrogels that can largely recapture the microenvironment of native stem cells. (3) Suggest a few future directions that can be implemented to improve current in vitro stem cell expansion systems.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"126085469","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 30
Harnessing Biomaterials and the Lymphatic System for Immunomodulation 利用生物材料和淋巴系统进行免疫调节
Pub Date : 2020-12-18 DOI: 10.2139/ssrn.3751564
Laura Alderfer, Eva Hall, Donny Hanjaya-Putra
The lymphatic system plays an integral part in regulating immune cells trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than the vascular system. To bridge the knowledge gap, biomaterials can be used to investigate the lymphatic system and to provide novel understanding into complex disease states, including cancer metastasis and inflammatory diseases. Insight gained from these mechanistic studies, can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration.  This review article focuses on recent progresses in (i) biomaterials used for lymphatic vessel formation, (ii) lymphatic models for studying lymphatic-immune cells interactions, (iii) pharmaceuticals and their interactions with the lymphatic system, (iv) drug screening, and (v) disease prediction. Finally, a number of challenges in adopting biomaterials for immunomodulation and future perspectives are discussed.
淋巴系统在调节免疫细胞的运输和大分子的运输中起着不可或缺的作用。然而,它对疾病进展和药物摄取的影响比血管系统了解得少。为了弥补知识差距,生物材料可以用于研究淋巴系统,并为复杂的疾病状态提供新的理解,包括癌症转移和炎症性疾病。从这些机制研究中获得的见解,可以进一步用于设计创新的生物材料来调节免疫系统,改善药物输送,促进组织再生。本文综述了以下方面的最新进展:(i)用于淋巴血管形成的生物材料,(ii)用于研究淋巴-免疫细胞相互作用的淋巴模型,(iii)药物及其与淋巴系统的相互作用,(iv)药物筛选,(v)疾病预测。最后,讨论了采用生物材料进行免疫调节的一些挑战和未来的展望。
{"title":"Harnessing Biomaterials and the Lymphatic System for Immunomodulation","authors":"Laura Alderfer, Eva Hall, Donny Hanjaya-Putra","doi":"10.2139/ssrn.3751564","DOIUrl":"https://doi.org/10.2139/ssrn.3751564","url":null,"abstract":"The lymphatic system plays an integral part in regulating immune cells trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than the vascular system. To bridge the knowledge gap, biomaterials can be used to investigate the lymphatic system and to provide novel understanding into complex disease states, including cancer metastasis and inflammatory diseases. Insight gained from these mechanistic studies, can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration.  This review article focuses on recent progresses in (i) biomaterials used for lymphatic vessel formation, (ii) lymphatic models for studying lymphatic-immune cells interactions, (iii) pharmaceuticals and their interactions with the lymphatic system, (iv) drug screening, and (v) disease prediction. Finally, a number of challenges in adopting biomaterials for immunomodulation and future perspectives are discussed.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"131836523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Imprinted Hydrogels with LbL Coating for Dual Drug Release from Soft Contact Lenses Materials 带有LbL涂层的印迹水凝胶用于软性隐形眼镜材料的双重药物释放
Pub Date : 2020-06-03 DOI: 10.2139/ssrn.3606830
Diana Silva, Hermínio C. de Sousa, Maria Helena Gil, Luís F. Santos, R. Amaral, Jorge A. Saraiva, M. Oom, C. Alvarez‐Lorenzo, A. P. Serro, B. Saramago
A combined strategy to control the release of two drugs, one anti-inflammatory (diclofenac sodium, DCF) and one antibiotic (moxifloxacin hydrochloride, MXF), from a soft contact lens (SCL) material, was assessed. The material was a silicone-based hydrogel, which was modified by molecular imprinting with MXF and coated by the layer-by-layer (LbL) method using natural polyelectrolytes: alginate (ALG), poly-l-lysine (PLL) and hyaluronate (HA), crosslinked with 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC). Imprinting was used to increase the amount of MXF loaded and to sustain its release, while the LbL coating acted as a diffusion barrier for DCF and improved the surface properties. The drugs were loaded by soaking in a DCF + MXF dual solution. High hydrostatic pressure (HHP) was successfully applied in the sterilization of the drug-loaded hydrogels. The transmittance, refractive index, wettability and ionic permeability of the hydrogels remained within the required levels for SCLs application. The concentrations of the released DCF and MXF stayed above the IC50 and the MIC (for S. aureus and S. epidermidis) values, for 9 and 10 days, respectively. No ocular irritancy was detected by the HET-CAM test. NIH/3T3 cell viability demonstrated that the drug-loaded hydrogels were not toxic, and cell adhesion was reduced.
评估了一种控制两种药物释放的联合策略,一种抗炎药物(双氯芬酸钠,DCF)和一种抗生素(盐酸莫西沙星,MXF)从软性隐形眼镜(SCL)材料中释放。该材料为硅基水凝胶,采用MXF分子印迹法对其进行修饰,并用海藻酸盐(ALG)、聚赖氨酸(PLL)和透明质酸盐(HA)等天然聚电解质与1-乙基-3-(3-二甲氨基丙基)盐酸碳二亚胺(EDC)交联,通过层层(LbL)法制备。印迹可以增加MXF的负载量并维持其释放,而LbL涂层可以作为DCF的扩散屏障并改善表面性能。用DCF + MXF双溶液浸泡装药。高静水压力(HHP)成功地应用于载药水凝胶的灭菌。水凝胶的透光率、折射率、润湿性和离子渗透性均保持在scl应用所需的水平。释放的DCF和MXF浓度分别高于IC50和MIC(金黄色葡萄球菌和表皮葡萄球菌)值,持续9天和10天。HET-CAM试验未检测到眼部刺激。NIH/3T3细胞活力表明,载药水凝胶无毒,细胞黏附降低。
{"title":"Imprinted Hydrogels with LbL Coating for Dual Drug Release from Soft Contact Lenses Materials","authors":"Diana Silva, Hermínio C. de Sousa, Maria Helena Gil, Luís F. Santos, R. Amaral, Jorge A. Saraiva, M. Oom, C. Alvarez‐Lorenzo, A. P. Serro, B. Saramago","doi":"10.2139/ssrn.3606830","DOIUrl":"https://doi.org/10.2139/ssrn.3606830","url":null,"abstract":"A combined strategy to control the release of two drugs, one anti-inflammatory (diclofenac sodium, DCF) and one antibiotic (moxifloxacin hydrochloride, MXF), from a soft contact lens (SCL) material, was assessed. The material was a silicone-based hydrogel, which was modified by molecular imprinting with MXF and coated by the layer-by-layer (LbL) method using natural polyelectrolytes: alginate (ALG), poly-l-lysine (PLL) and hyaluronate (HA), crosslinked with 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC). Imprinting was used to increase the amount of MXF loaded and to sustain its release, while the LbL coating acted as a diffusion barrier for DCF and improved the surface properties. The drugs were loaded by soaking in a DCF + MXF dual solution. High hydrostatic pressure (HHP) was successfully applied in the sterilization of the drug-loaded hydrogels. The transmittance, refractive index, wettability and ionic permeability of the hydrogels remained within the required levels for SCLs application. The concentrations of the released DCF and MXF stayed above the IC50 and the MIC (for S. aureus and S. epidermidis) values, for 9 and 10 days, respectively. No ocular irritancy was detected by the HET-CAM test. NIH/3T3 cell viability demonstrated that the drug-loaded hydrogels were not toxic, and cell adhesion was reduced.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"122736485","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 18
Bifunctional Bio-Surface Decorated Bone-Grafting Titanium Material with Cancellous Bone-Like Biomimetic Structure for Enhanced Bone Tissue Regeneration 具有松质骨样仿生结构的双功能生物表面修饰植骨钛材料增强骨组织再生
Pub Date : 2020-04-29 DOI: 10.2139/ssrn.3577287
Bingjun Zhang, J. Li, Lei He, Hao Huang, J. Weng
In view of the fact that titanium (Ti)-based implants still face the problem of loosening and failure of the implants caused by the slow biological response, the low osseointegration rate and the implant bacterial infection in clinical application, we designed a cancellous bone-like biomimetic Ti scaffold via using the template accumulated by sugar spheres as pore-forming agent. And based on a modified surface mineralization process and mussel-like adhesion mechanism, a silicon-doped calcium phosphate composite coating (Van-pBNPs/pep@pSiCaP) with Vancomycin (Van)-loaded polydopamine (pDA)-modified albumin nanoparticles (Van-pBNPs) and cell adhesion peptides (GFOGER) was constructed on the surface of Ti scaffold for mimicking the extracellular matrix (ECM) microenvironment of natural bone matrix to induce greater tissue regeneration. The in vitro study demonstrated that this porous Ti scaffold with functional bio-surface could distinctly facilitate cell early adhesion and spreading, and activate the expression of α2β1 integrin receptor on the cell membrane through promoting the formation of focal adhesions (FAs) in bone marrow stromal cells (BMSCs), thus mediating greater osteogenic cell differentiation. And it could also effectively inhibit the adhesion and growth of Staphylococcus epidermidis, exhibiting excellent antibacterial properties. Moreover, the Van-pBNPs/pep@pSiCaP-Ti scaffolds showed enhanced in vivo bone-forming ability due to the contributions of bioactive chemical components and the natural cancellous bone-like macrostructure. This work offers a promising structural and functional bio-inspired strategy for designing metal implants with desirable ability of osteoinduction synergistically with antibacterial efficacy for promoting bone regeneration and infection prevention simultaneously.
鉴于钛(Ti)基种植体在临床应用中仍存在生物反应缓慢、骨整合率低、种植体细菌感染等导致种植体松动失效的问题,我们利用糖球积累的模板作为成孔剂,设计了一种松质骨样仿生钛支架。基于改良的表面矿化过程和贻贝样粘附机制,在Ti支架表面构建了载万古霉素(Van)修饰的聚多巴胺(pDA)修饰的白蛋白纳米颗粒(Van- pbnps)和细胞粘附肽(GFOGER)的掺硅磷酸钙复合涂层(Van- pbnps /pep@pSiCaP),以模拟天然骨基质的细胞外基质(ECM)微环境,诱导更大的组织再生。体外研究表明,这种具有功能性生物表面的多孔钛支架可以明显促进细胞早期粘附和扩散,并通过促进骨髓基质细胞(BMSCs)的局灶粘连(FAs)的形成,激活细胞膜上α2β1整合素受体的表达,从而介导更大的成骨细胞分化。并能有效抑制表皮葡萄球菌的粘附和生长,具有良好的抗菌性能。此外,由于生物活性化学成分和天然松质骨样宏观结构的贡献,Van-pBNPs/pep@pSiCaP-Ti支架具有增强的体内成骨能力。这项工作为设计具有理想的骨诱导能力的金属植入物提供了一种有前途的结构和功能的仿生策略,同时具有抗菌功效,促进骨再生和预防感染。
{"title":"Bifunctional Bio-Surface Decorated Bone-Grafting Titanium Material with Cancellous Bone-Like Biomimetic Structure for Enhanced Bone Tissue Regeneration","authors":"Bingjun Zhang, J. Li, Lei He, Hao Huang, J. Weng","doi":"10.2139/ssrn.3577287","DOIUrl":"https://doi.org/10.2139/ssrn.3577287","url":null,"abstract":"In view of the fact that titanium (Ti)-based implants still face the problem of loosening and failure of the implants caused by the slow biological response, the low osseointegration rate and the implant bacterial infection in clinical application, we designed a cancellous bone-like biomimetic Ti scaffold via using the template accumulated by sugar spheres as pore-forming agent. And based on a modified surface mineralization process and mussel-like adhesion mechanism, a silicon-doped calcium phosphate composite coating (Van-pBNPs/pep@pSiCaP) with Vancomycin (Van)-loaded polydopamine (pDA)-modified albumin nanoparticles (Van-pBNPs) and cell adhesion peptides (GFOGER) was constructed on the surface of Ti scaffold for mimicking the extracellular matrix (ECM) microenvironment of natural bone matrix to induce greater tissue regeneration. The in vitro study demonstrated that this porous Ti scaffold with functional bio-surface could distinctly facilitate cell early adhesion and spreading, and activate the expression of α2β1 integrin receptor on the cell membrane through promoting the formation of focal adhesions (FAs) in bone marrow stromal cells (BMSCs), thus mediating greater osteogenic cell differentiation. And it could also effectively inhibit the adhesion and growth of Staphylococcus epidermidis, exhibiting excellent antibacterial properties. Moreover, the Van-pBNPs/pep@pSiCaP-Ti scaffolds showed enhanced in vivo bone-forming ability due to the contributions of bioactive chemical components and the natural cancellous bone-like macrostructure. This work offers a promising structural and functional bio-inspired strategy for designing metal implants with desirable ability of osteoinduction synergistically with antibacterial efficacy for promoting bone regeneration and infection prevention simultaneously.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"123894380","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Self-Assembled Polydopamine Nanoparticles Improve Treatment in Parkinson's Disease Model Mice and Suppress Dopamine-Induced Dyskinesia 自组装聚多巴胺纳米颗粒改善帕金森病模型小鼠的治疗并抑制多巴胺诱导的运动障碍
Pub Date : 2020-02-21 DOI: 10.2139/ssrn.3539216
L. Vong, Yuna Sato, Pennapa Chonpathompikunlert, Supita Tanasawet, P. Hutamekalin, Y. Nagasaki
Although Levodopa (L-DOPA), a dopamine precursor, exhibits a high risk of dyskinesia, it remains the primary treatment in Parkinson's disease (PD), a progressive neurodegenerative disorder. In this study, we designed poly(L-DOPA)-based self-assembled nanodrug (NanoDOPA) from amphiphilic block copolymer possessing poly(L-DOPA(OAc)2), which is a precursor of L-DOPA as a hydrophobic segment, for treatment in a PD model mouse. Under physiological enzyme treatment, the poly(L-DOPA(OAc)2) in the block copolymer was hydrolyzed to liberate L-DOPA gradually. Using the MPTP-induced PD mouse model, we observed that mice treated with NanoDOPA demonstrated a significant improvement of PD symptoms compared to the L-DOPA treatment. Interestingly, the NanoDOPA treatment did not cause the dyskinesia symptoms, which was clearly observed in the L-DOPA-treated mice. Furthermore, NanoDOPA exhibited remarkably lower toxicity in vitro compared to L-DOPA, in addition with no noticeable NanoDOPA toxicity observed in the treated mice. These results suggested that self-assembled NanoDOPA is a promising therapeutic in the treatment of PD. Statement of significance In this study, we proposed a therapeutic approach for the effective treatment of Parkinson disease (PD) using newly designed poly(L-DOPA)-based self-assembled nanodrug (NanoDOPA) prepared from amphiphilic block copolymers possessing poly(L-DOPA(OAc)2), which is a precursor of L-DOPA as a hydrophobic segment, for treatment in a PD model mouse. Under physiological enzyme treatments, NanoDOPA was hydrolyzed to liberate L-DOPA gradually, improving the pharmacokinetic value of L-DOPA. The mice treated with NanoDOPA significantly improved PD symptoms compared to the L-DOPA treatment in a neurotoxin-induced PD mouse model. Interestingly, NanoDOPA treatment did not cause dyskinesia symptoms, which was observed in the L-DOPA-treated mice. The obtained results in this study suggested that self-assembly NanoDOPA is a promising therapeutic in the treatment of PD.
尽管多巴胺前体左旋多巴(L-DOPA)具有较高的运动障碍风险,但它仍然是帕金森病(一种进行性神经退行性疾病)的主要治疗方法。在本研究中,我们设计了基于聚(L-DOPA)的自组装纳米药物(NanoDOPA),由两亲嵌段共聚物含有聚(L-DOPA(OAc)2),这是L-DOPA的前体作为疏水段,用于治疗PD模型小鼠。在生理酶处理下,嵌段共聚物中的聚L-DOPA(OAc)2)被水解,逐渐释放出L-DOPA。使用mptp诱导的PD小鼠模型,我们观察到与左旋多巴治疗相比,纳米多巴治疗的小鼠PD症状有显著改善。有趣的是,NanoDOPA治疗并没有引起运动障碍症状,这在左旋多巴治疗的小鼠中可以清楚地观察到。此外,与左旋多巴相比,NanoDOPA在体外表现出明显较低的毒性,此外在处理小鼠中未观察到明显的NanoDOPA毒性。这些结果表明,自组装纳米多巴是一种很有前景的治疗PD的药物。在这项研究中,我们提出了一种有效治疗帕金森病(PD)的方法,该方法是由含有聚(L-DOPA(OAc)2)的两亲嵌段共聚物制备的基于聚(L-DOPA)的自组装纳米药物(NanoDOPA),它是L-DOPA的前体作为疏水段,用于治疗PD模型小鼠。在生理酶处理下,NanoDOPA被水解逐渐释放出左旋多巴,提高了左旋多巴的药动学值。在神经毒素诱导的帕金森小鼠模型中,与左旋多巴治疗相比,用纳米多巴治疗的小鼠帕金森症状明显改善。有趣的是,纳米多巴治疗没有引起运动障碍症状,这在左旋多巴治疗的小鼠中观察到。本研究结果表明,自组装纳米多巴是一种很有前景的治疗PD的药物。
{"title":"Self-Assembled Polydopamine Nanoparticles Improve Treatment in Parkinson's Disease Model Mice and Suppress Dopamine-Induced Dyskinesia","authors":"L. Vong, Yuna Sato, Pennapa Chonpathompikunlert, Supita Tanasawet, P. Hutamekalin, Y. Nagasaki","doi":"10.2139/ssrn.3539216","DOIUrl":"https://doi.org/10.2139/ssrn.3539216","url":null,"abstract":"Although Levodopa (L-DOPA), a dopamine precursor, exhibits a high risk of dyskinesia, it remains the primary treatment in Parkinson's disease (PD), a progressive neurodegenerative disorder. In this study, we designed poly(L-DOPA)-based self-assembled nanodrug (NanoDOPA) from amphiphilic block copolymer possessing poly(L-DOPA(OAc)2), which is a precursor of L-DOPA as a hydrophobic segment, for treatment in a PD model mouse. Under physiological enzyme treatment, the poly(L-DOPA(OAc)2) in the block copolymer was hydrolyzed to liberate L-DOPA gradually. Using the MPTP-induced PD mouse model, we observed that mice treated with NanoDOPA demonstrated a significant improvement of PD symptoms compared to the L-DOPA treatment. Interestingly, the NanoDOPA treatment did not cause the dyskinesia symptoms, which was clearly observed in the L-DOPA-treated mice. Furthermore, NanoDOPA exhibited remarkably lower toxicity in vitro compared to L-DOPA, in addition with no noticeable NanoDOPA toxicity observed in the treated mice. These results suggested that self-assembled NanoDOPA is a promising therapeutic in the treatment of PD. Statement of significance In this study, we proposed a therapeutic approach for the effective treatment of Parkinson disease (PD) using newly designed poly(L-DOPA)-based self-assembled nanodrug (NanoDOPA) prepared from amphiphilic block copolymers possessing poly(L-DOPA(OAc)2), which is a precursor of L-DOPA as a hydrophobic segment, for treatment in a PD model mouse. Under physiological enzyme treatments, NanoDOPA was hydrolyzed to liberate L-DOPA gradually, improving the pharmacokinetic value of L-DOPA. The mice treated with NanoDOPA significantly improved PD symptoms compared to the L-DOPA treatment in a neurotoxin-induced PD mouse model. Interestingly, NanoDOPA treatment did not cause dyskinesia symptoms, which was observed in the L-DOPA-treated mice. The obtained results in this study suggested that self-assembly NanoDOPA is a promising therapeutic in the treatment of PD.","PeriodicalId":105746,"journal":{"name":"AMI: Acta Biomaterialia","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"129224660","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 32
期刊
AMI: Acta Biomaterialia
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