首页 > 最新文献

Biofabrication最新文献

英文 中文
Endothelial extracellular vesicles enhance vascular self-assembly in engineered human cardiac tissues. 内皮细胞外囊泡增强了工程人体心脏组织的血管自组装能力。
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-09-03 DOI: 10.1088/1758-5090/ad76d9
Karl T Wagner, Rick X Z Lu, Shira Landau, Sarah A Shawky, Yimu Zhao, David F Bodenstein, Luis Felipe Jiménez Vargas, Richard Jiang, Sargol Okhovatian, Ying Wang, Chuan Liu, Daniel Vosoughi, Dakota Gustafson, Jason E Fish, Carolyn L Cummins, Milica Radisic

The fabrication of complex and stable vasculature in engineered cardiac tissues represents a significant hurdle towards building physiologically relevant models of the heart. Here, we implemented a 3D model of cardiac vasculogenesis, incorporating endothelial cells (EC), stromal cells, and human iPSC-derived cardiomyocytes (CM) in a fibrin hydrogel. The presence of CMs disrupted vessel formation in 3D tissues, resulting in the upregulation of endothelial activation markers and altered extracellular vesicle (EV) signaling in engineered tissues as determined by the proteomic analysis of culture supernatant. miRNA sequencing of CM- and EC-secreted EVs highlighted key EV-miRNAs that were postulated to play differing roles in cardiac vasculogenesis, including the let-7 family and miR-126-3p in EC-EVs. In the absence of CMs, the supplementation of CM-EVs to EC monolayers attenuated EC migration and proliferation and resulted in shorter and more discontinuous self-assembling vessels when applied to 3D vascular tissues. In contrast, supplementation of EC-EVs to the tissue culture media of 3D vascularized cardiac tissues mitigated some of the deleterious effects of CMs on vascular self-assembly, enhancing the average length and continuity of vessel tubes that formed in the presence of CMs. Direct transfection validated the effects of the key EC-EV miRNAs let-7b-5p and miR-126-3p in improving the maintenance of continuous vascular networks. EC-EV supplementation to biofabricated cardiac tissues and microfluidic devices resulted in tissue vascularization, illustrating the use of this approach in the engineering of enhanced, perfusable, microfluidic models of the myocardium.

在工程心脏组织中制造复杂而稳定的血管是建立与生理相关的心脏模型的一大障碍。在这里,我们在纤维蛋白水凝胶中加入了内皮细胞(EC)、基质细胞和人类 iPSC 衍生心肌细胞(CM),实现了心脏血管生成的三维模型。CM的存在破坏了三维组织中的血管形成,导致内皮活化标志物上调,并改变了工程组织中的细胞外囊泡(EV)信号,这是由培养上清液的蛋白质组分析确定的。在没有CMs的情况下,将CM-EVs补充到EC单层中会减少EC的迁移和增殖,并在应用于三维血管组织时导致更短和更不连续的自组装血管。与此相反,在三维血管化心脏组织的组织培养基中添加 EC-EVs 可减轻 CMs 对血管自组装的一些有害影响,增强在 CMs 存在下形成的血管管的平均长度和连续性。直接转染验证了关键的 EC-EV miRNA let-7b-5p 和 miR-126-3p 在改善连续血管网络的维持方面的作用。向生物制造的心脏组织和微流控装置补充 EC-EV 可实现组织血管化,这说明这种方法可用于制造增强的、可灌注的心肌微流控模型。
{"title":"Endothelial extracellular vesicles enhance vascular self-assembly in engineered human cardiac tissues.","authors":"Karl T Wagner, Rick X Z Lu, Shira Landau, Sarah A Shawky, Yimu Zhao, David F Bodenstein, Luis Felipe Jiménez Vargas, Richard Jiang, Sargol Okhovatian, Ying Wang, Chuan Liu, Daniel Vosoughi, Dakota Gustafson, Jason E Fish, Carolyn L Cummins, Milica Radisic","doi":"10.1088/1758-5090/ad76d9","DOIUrl":"https://doi.org/10.1088/1758-5090/ad76d9","url":null,"abstract":"<p><p>The fabrication of complex and stable vasculature in engineered cardiac tissues represents a significant hurdle towards building physiologically relevant models of the heart. Here, we implemented a 3D model of cardiac vasculogenesis, incorporating endothelial cells (EC), stromal cells, and human iPSC-derived cardiomyocytes (CM) in a fibrin hydrogel. The presence of CMs disrupted vessel formation in 3D tissues, resulting in the upregulation of endothelial activation markers and altered extracellular vesicle (EV) signaling in engineered tissues as determined by the proteomic analysis of culture supernatant. miRNA sequencing of CM- and EC-secreted EVs highlighted key EV-miRNAs that were postulated to play differing roles in cardiac vasculogenesis, including the let-7 family and miR-126-3p in EC-EVs. In the absence of CMs, the supplementation of CM-EVs to EC monolayers attenuated EC migration and proliferation and resulted in shorter and more discontinuous self-assembling vessels when applied to 3D vascular tissues. In contrast, supplementation of EC-EVs to the tissue culture media of 3D vascularized cardiac tissues mitigated some of the deleterious effects of CMs on vascular self-assembly, enhancing the average length and continuity of vessel tubes that formed in the presence of CMs. Direct transfection validated the effects of the key EC-EV miRNAs let-7b-5p and miR-126-3p in improving the maintenance of continuous vascular networks. EC-EV supplementation to biofabricated cardiac tissues and microfluidic devices resulted in tissue vascularization, illustrating the use of this approach in the engineering of enhanced, perfusable, microfluidic models of the myocardium.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124728","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effect of viscosity of gelatin methacryloyl-based bioinks on bone cells. 明胶甲基丙烯酰基生物墨水的粘度对骨细胞的影响
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-09-03 DOI: 10.1088/1758-5090/ad6d91
Ahmad Rashad, Alejandro Gomez, Ankit Gangrade, Fatemeh Zehtabi, Kalpana Mandal, Surjendu Maity, Changyu Ma, Bingbing Li, Ali Khademhosseini, Natan Roberto de Barros

The viscosity of gelatin methacryloyl (GelMA)-based bioinks generates shear stresses throughout the printing process that can affect cell integrity, reduce cell viability, cause morphological changes, and alter cell functionality. This study systematically investigated the impact of the viscosity of GelMA-gelatin bioinks on osteoblast-like cells in 2D and 3D culture conditions. Three bioinks with low, medium, and high viscosity prepared by supplementing a 5% GelMA solution with different concentrations of gelatin were evaluated. Cell responses were studied in a 2D environment after printing and incubation in non-cross-linked bioinks that caused the gelatin and GelMA to dissolve and release cells for attachment to tissue culture plates. The increased viscosity of the bioinks significantly affected cell area and aspect ratio. Cells printed using the bioink with medium viscosity exhibited greater metabolic activity and proliferation rate than those printed using the high viscosity bioink and even the unprinted control cells. Additionally, cells printed using the bioink with high viscosity demonstrated notably elevated expression levels of alkaline phosphatase and bone morphogenetic protein-2 genes. In the 3D condition, the printed cell-laden hydrogels were photo-cross-linked prior to incubation. The medium viscosity bioink supported greater cell proliferation compared to the high viscosity bioink. However, there were no significant differences in the expression of osteogenic markers between the medium and high viscosity bioinks. Therefore, the choice between medium and high viscosity bioinks should be based on the desired outcomes and objectives of the bone tissue engineering application. Furthermore, the bioprinting procedure with the medium viscosity bioink was used as an automated technique for efficiently seeding cells onto 3D printed porous titanium scaffolds for bone tissue engineering purposes.

明胶甲基丙烯酰(GelMA)基生物墨水的粘度会在整个打印过程中产生剪切应力,从而影响细胞的完整性、降低细胞活力、导致形态变化并改变细胞功能。本研究系统地研究了在二维和三维培养条件下,GelMA-明胶生物墨水的粘度对类成骨细胞的影响。通过在 5% 的 GelMA 溶液中添加不同浓度的明胶,制备出低、中、高粘度的三种生物水墨,并对其进行了评估。研究了在二维环境中打印和在非交联生物墨水中培养后的细胞反应,这些生物墨水可使明胶和 GelMA 溶解并释放细胞,使细胞附着在组织培养板上。生物墨水粘度的增加极大地影响了细胞面积和长宽比。与使用高粘度生物墨水打印的细胞甚至未打印的对照细胞相比,使用中等粘度生物墨水打印的细胞表现出更高的代谢活性和增殖率。此外,使用高粘度生物墨水打印的细胞,其碱性磷酸酶(ALP)和骨形态发生蛋白-2(BMP-2)基因的表达水平明显升高。在三维条件下,打印出的含有细胞的水凝胶在培养前进行了光交联。与高粘度生物墨水相比,中等粘度生物墨水支持更多的细胞增殖。然而,中粘度和高粘度生物墨水在成骨标志物的表达上没有明显差异。因此,应根据骨组织工程应用的预期结果和目标来选择中粘度和高粘度生物墨水。此外,使用中等粘度生物墨水的生物打印程序被用作一种自动化技术,可有效地将细胞播种到用于骨组织工程的三维打印多孔钛支架上。
{"title":"Effect of viscosity of gelatin methacryloyl-based bioinks on bone cells.","authors":"Ahmad Rashad, Alejandro Gomez, Ankit Gangrade, Fatemeh Zehtabi, Kalpana Mandal, Surjendu Maity, Changyu Ma, Bingbing Li, Ali Khademhosseini, Natan Roberto de Barros","doi":"10.1088/1758-5090/ad6d91","DOIUrl":"10.1088/1758-5090/ad6d91","url":null,"abstract":"<p><p>The viscosity of gelatin methacryloyl (GelMA)-based bioinks generates shear stresses throughout the printing process that can affect cell integrity, reduce cell viability, cause morphological changes, and alter cell functionality. This study systematically investigated the impact of the viscosity of GelMA-gelatin bioinks on osteoblast-like cells in 2D and 3D culture conditions. Three bioinks with low, medium, and high viscosity prepared by supplementing a 5% GelMA solution with different concentrations of gelatin were evaluated. Cell responses were studied in a 2D environment after printing and incubation in non-cross-linked bioinks that caused the gelatin and GelMA to dissolve and release cells for attachment to tissue culture plates. The increased viscosity of the bioinks significantly affected cell area and aspect ratio. Cells printed using the bioink with medium viscosity exhibited greater metabolic activity and proliferation rate than those printed using the high viscosity bioink and even the unprinted control cells. Additionally, cells printed using the bioink with high viscosity demonstrated notably elevated expression levels of alkaline phosphatase and bone morphogenetic protein-2 genes. In the 3D condition, the printed cell-laden hydrogels were photo-cross-linked prior to incubation. The medium viscosity bioink supported greater cell proliferation compared to the high viscosity bioink. However, there were no significant differences in the expression of osteogenic markers between the medium and high viscosity bioinks. Therefore, the choice between medium and high viscosity bioinks should be based on the desired outcomes and objectives of the bone tissue engineering application. Furthermore, the bioprinting procedure with the medium viscosity bioink was used as an automated technique for efficiently seeding cells onto 3D printed porous titanium scaffolds for bone tissue engineering purposes.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911582","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
High-yield extracellular vesicle production from HEK293T cells encapsulated in 3D auxetic scaffolds with cyclic mechanical stimulation for effective drug carrier systems. 在三维辅助支架中封装的 HEK293T 细胞在循环机械刺激下高产产生细胞外囊泡,从而形成有效的药物载体系统。
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-09-02 DOI: 10.1088/1758-5090/ad728b
Yi-Wen Chen, Yen-Hong Lin, Chia-Che Ho, Cheng-Yu Chen, Min-Hua Yu, Alvin Kai-Xing Lee, Shao-Chih Chiu, Der-Yang Cho, Ming-You Shie

Extracellular vesicles (EVs) show promise in drug loading and delivery for medical applications. However, the lack of scalable manufacturing processes hinders the generation of clinically suitable quantities, thereby impeding the translation of EV-based therapies. Current EV production relies heavily on non-physiological two-dimensional (2D) cell culture or bioreactors, requiring significant resources. Additionally, EV-derived ribonucleic acid cargo in three-dimensional (3D) and 2D culture environments remains largely unknown. In this study, we optimized the biofabrication of 3D auxetic scaffolds encapsulated with human embryonic kidney 293 T (HEK293 T) cells, focusing on enhancing the mechanical properties of the scaffolds to significantly boost EV production through tensile stimulation in bioreactors. The proposed platform increased EV yields approximately 115-fold compared to conventional 2D culture, possessing properties that inhibit tumor progression. Further mechanistic examinations revealed that this effect was mediated by the mechanosensitivity of YAP/TAZ. EVs derived from tensile-stimulated HEK293 T cells on 3D auxetic scaffolds demonstrated superior capability for loading doxorubicin compared to their 2D counterparts for cancer therapy. Our results underscore the potential of this strategy for scaling up EV production and optimizing functional performance for clinical translation.

细胞外囊泡(EVs)在医疗应用中显示出装载和输送药物的前景。然而,由于缺乏可扩展的生产工艺,无法生产出临床适用的数量,从而阻碍了基于 EV 的疗法的转化。目前的 EV 生产严重依赖非生理性的二维细胞培养或生物反应器,需要大量资源。此外,在三维和二维培养环境中,EV 衍生的核糖核酸货物在很大程度上仍然未知。在本研究中,我们优化了包裹有人类胚胎肾脏 293T(HEK293T)细胞的三维辅助支架的生物制造,重点是增强支架的机械性能,通过生物反应器中的拉伸刺激显著提高 EV 产量。与传统的二维培养相比,所提出的平台使EV产量提高了约115倍,并具有抑制肿瘤进展的特性。进一步的机理研究发现,这种效应是由 YAP/TAZ 的机械敏感性介导的。三维辅助支架上的 HEK293T 细胞在拉伸刺激下产生的 EVs 在负载多柔比星方面的能力优于二维EVs。我们的研究结果凸显了这一策略在扩大 EV 生产规模和优化临床转化功能性能方面的潜力。
{"title":"High-yield extracellular vesicle production from HEK293T cells encapsulated in 3D auxetic scaffolds with cyclic mechanical stimulation for effective drug carrier systems.","authors":"Yi-Wen Chen, Yen-Hong Lin, Chia-Che Ho, Cheng-Yu Chen, Min-Hua Yu, Alvin Kai-Xing Lee, Shao-Chih Chiu, Der-Yang Cho, Ming-You Shie","doi":"10.1088/1758-5090/ad728b","DOIUrl":"10.1088/1758-5090/ad728b","url":null,"abstract":"<p><p>Extracellular vesicles (EVs) show promise in drug loading and delivery for medical applications. However, the lack of scalable manufacturing processes hinders the generation of clinically suitable quantities, thereby impeding the translation of EV-based therapies. Current EV production relies heavily on non-physiological two-dimensional (2D) cell culture or bioreactors, requiring significant resources. Additionally, EV-derived ribonucleic acid cargo in three-dimensional (3D) and 2D culture environments remains largely unknown. In this study, we optimized the biofabrication of 3D auxetic scaffolds encapsulated with human embryonic kidney 293 T (HEK293 T) cells, focusing on enhancing the mechanical properties of the scaffolds to significantly boost EV production through tensile stimulation in bioreactors. The proposed platform increased EV yields approximately 115-fold compared to conventional 2D culture, possessing properties that inhibit tumor progression. Further mechanistic examinations revealed that this effect was mediated by the mechanosensitivity of YAP/TAZ. EVs derived from tensile-stimulated HEK293 T cells on 3D auxetic scaffolds demonstrated superior capability for loading doxorubicin compared to their 2D counterparts for cancer therapy. Our results underscore the potential of this strategy for scaling up EV production and optimizing functional performance for clinical translation.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142035129","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
3Din vitromodeling of the exocrine pancreatic unit using tomographic volumetric bioprinting. 利用断层体积生物打印技术对胰腺外分泌单元进行三维虚拟建模。
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-08-30 DOI: 10.1088/1758-5090/ad6d8d
Viola Sgarminato, Jorge Madrid-Wolff, Antoine Boniface, Gianluca Ciardelli, Chiara Tonda-Turo, Christophe Moser

Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, a leading cause of cancer-related deaths globally. Initial lesions of PDAC develop within the exocrine pancreas' functional units, with tumor progression driven by interactions between PDAC and stromal cells. Effective therapies require anatomically and functionally relevantin vitrohuman models of the pancreatic cancer microenvironment. We employed tomographic volumetric bioprinting, a novel biofabrication method, to create human fibroblast-laden constructs mimicking the tubuloacinar structures of the exocrine pancreas. Human pancreatic ductal epithelial (HPDE) cells overexpressing the KRAS oncogene (HPDE-KRAS) were seeded in the multiacinar cavity to replicate pathological tissue. HPDE cell growth and organization within the structure were assessed, demonstrating the formation of a thin epithelium covering the acini inner surfaces. Immunofluorescence assays showed significantly higher alpha smooth muscle actin (α-SMA) vs. F-actin expression in fibroblasts co-cultured with cancerous versus wild-type HPDE cells. Additionally,α-SMA expression increased over time and was higher in fibroblasts closer to HPDE cells. Elevated interleukin (IL)-6 levels were quantified in supernatants from co-cultures of stromal and HPDE-KRAS cells. These findings align with inflamed tumor-associated myofibroblast behavior, serving as relevant biomarkers to monitor early disease progression and target drug efficacy. To our knowledge, this is the first demonstration of a 3D bioprinted model of exocrine pancreas that recapitulates its true 3-dimensional microanatomy and shows tumor triggered inflammation.

胰腺导管腺癌(PDAC)是最常见的胰腺癌类型,也是全球癌症相关死亡的主要原因。PDAC 最初的病变发生在胰腺外分泌功能单元内,PDAC 和基质细胞之间的相互作用推动了肿瘤的发展。有效的疗法需要与胰腺癌微环境解剖和功能相关的体外人体模型。我们采用断层容积生物打印这种新型生物制造方法,制造出了模拟胰腺外分泌管状结构的充满成纤维细胞的人体构建体。将过表达 KRAS 癌基因(HPDE-KRAS)的人胰腺导管上皮(HPDE)细胞播种到多囊腔中,以复制病理组织。评估了HPDE细胞在该结构中的生长和组织情况,结果显示形成了覆盖在尖头内表面的薄上皮。免疫荧光测定显示,与癌细胞共同培养的成纤维细胞中,α-平滑肌肌动蛋白(α-SMA)与F-肌动蛋白的表达明显高于野生型HPDE细胞。此外,α-SMA的表达随着时间的推移而增加,在更接近HPDE细胞的成纤维细胞中表达更高。在基质细胞和 HPDE-KRAS 细胞共培养的上清液中,白细胞介素 (IL)-6 水平升高。这些发现与发炎的肿瘤相关肌成纤维细胞行为一致,可作为监测早期疾病进展和靶向药物疗效的相关生物标记物。据我们所知,这是首次展示三维生物打印的外分泌胰腺模型,它再现了胰腺的真实三维微观解剖,并显示了肿瘤引发的炎症。
{"title":"3D<i>in vitro</i>modeling of the exocrine pancreatic unit using tomographic volumetric bioprinting.","authors":"Viola Sgarminato, Jorge Madrid-Wolff, Antoine Boniface, Gianluca Ciardelli, Chiara Tonda-Turo, Christophe Moser","doi":"10.1088/1758-5090/ad6d8d","DOIUrl":"10.1088/1758-5090/ad6d8d","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, a leading cause of cancer-related deaths globally. Initial lesions of PDAC develop within the exocrine pancreas' functional units, with tumor progression driven by interactions between PDAC and stromal cells. Effective therapies require anatomically and functionally relevant<i>in vitro</i>human models of the pancreatic cancer microenvironment. We employed tomographic volumetric bioprinting, a novel biofabrication method, to create human fibroblast-laden constructs mimicking the tubuloacinar structures of the exocrine pancreas. Human pancreatic ductal epithelial (HPDE) cells overexpressing the KRAS oncogene (HPDE-KRAS) were seeded in the multiacinar cavity to replicate pathological tissue. HPDE cell growth and organization within the structure were assessed, demonstrating the formation of a thin epithelium covering the acini inner surfaces. Immunofluorescence assays showed significantly higher alpha smooth muscle actin (<i>α</i>-SMA) vs. F-actin expression in fibroblasts co-cultured with cancerous versus wild-type HPDE cells. Additionally,<i>α</i>-SMA expression increased over time and was higher in fibroblasts closer to HPDE cells. Elevated interleukin (IL)-6 levels were quantified in supernatants from co-cultures of stromal and HPDE-KRAS cells. These findings align with inflamed tumor-associated myofibroblast behavior, serving as relevant biomarkers to monitor early disease progression and target drug efficacy. To our knowledge, this is the first demonstration of a 3D bioprinted model of exocrine pancreas that recapitulates its true 3-dimensional microanatomy and shows tumor triggered inflammation.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911580","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Vascular tissues bioprinted with smooth muscle cell-only bioinks in support baths mimic features of native coronary arteries. 在支撑浴中使用纯平滑肌细胞生物墨水进行生物打印的血管组织模仿了原生冠状动脉的特征。
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-08-29 DOI: 10.1088/1758-5090/ad6d8f
Andre E Figueroa-Milla, William DeMaria, Derrick Wells, Oju Jeon, Eben Alsberg, Marsha W Rolle

This study explores the bioprinting of a smooth muscle cell-only bioink into ionically crosslinked oxidized methacrylated alginate (OMA) microgel baths to create self-supporting vascular tissues. The impact of OMA microgel support bath methacrylation degree and cell-only bioink dispensing parameters on tissue formation, remodeling, structure and strength was investigated. We hypothesized that reducing dispensing tip diameter from 27 G (210μm) to 30 G (159μm) for cell-only bioink dispensing would reduce tissue wall thickness and improve the consistency of tissue dimensions while maintaining cell viability. Printing with 30 G tips resulted in decreased mean wall thickness (318.6μm) without compromising mean cell viability (94.8%). Histological analysis of cell-only smooth muscle tissues cultured for 14 d in OMA support baths exhibited decreased wall thickness using 30 G dispensing tips, which correlated with increased collagen deposition and alignment. In addition, a TUNEL assay indicated a decrease in cell death in tissues printed with thinner (30 G) dispensing tips. Mechanical testing demonstrated that tissues printed with a 30 G dispensing tip exhibit an increase in ultimate tensile strength compared to those printed with a 27 G dispensing tip. Overall, these findings highlight the importance of precise control over bioprinting parameters to generate mechanically robust tissues when using cell-only bioinks dispensed and cultured within hydrogel support baths. The ability to control print dimensions using cell-only bioinks may enable bioprinting of more complex soft tissue geometries to generatein vitrotissue models.

本研究探讨了将纯平滑肌细胞生物墨水注入离子交联氧化甲基丙烯酸海藻酸盐(OMA)微凝胶浴中进行生物打印,以创建自支撑血管组织的方法。我们研究了 OMA 微凝胶支撑浴甲基丙烯酸化程度和纯细胞生物墨水分配参数对组织形成、重塑、结构和强度的影响。我们假设,将纯细胞生物墨水点胶时的点胶头直径从 27G(210 微米)减小到 30G(159 微米),可以在保持细胞活力的同时,减少组织壁厚度,提高组织尺寸的一致性。使用 30G 针尖打印可减少平均壁厚(318.6 微米),而不影响平均细胞存活率(94.8%)。在 OMA 支持浴中培养 14 天的纯细胞平滑肌组织的组织学分析表明,使用 30G 分配吸头可减少壁厚,这与胶原沉积和排列增加有关。此外,TUNEL 检测表明,使用较细(30G)点胶头打印的组织细胞死亡减少。机械测试表明,与使用 27G 点胶针头打印的组织相比,使用 30G 点胶针头打印的组织的极限拉伸强度有所提高。总之,这些发现强调了精确控制生物打印参数的重要性,以便在水凝胶支撑槽中使用纯细胞生物墨水点胶和培养时生成机械强度高的组织。使用纯细胞生物墨水控制打印尺寸的能力可实现更复杂软组织几何形状的生物打印,从而生成体外组织模型。
{"title":"Vascular tissues bioprinted with smooth muscle cell-only bioinks in support baths mimic features of native coronary arteries.","authors":"Andre E Figueroa-Milla, William DeMaria, Derrick Wells, Oju Jeon, Eben Alsberg, Marsha W Rolle","doi":"10.1088/1758-5090/ad6d8f","DOIUrl":"10.1088/1758-5090/ad6d8f","url":null,"abstract":"<p><p>This study explores the bioprinting of a smooth muscle cell-only bioink into ionically crosslinked oxidized methacrylated alginate (OMA) microgel baths to create self-supporting vascular tissues. The impact of OMA microgel support bath methacrylation degree and cell-only bioink dispensing parameters on tissue formation, remodeling, structure and strength was investigated. We hypothesized that reducing dispensing tip diameter from 27 G (210<i>μ</i>m) to 30 G (159<i>μ</i>m) for cell-only bioink dispensing would reduce tissue wall thickness and improve the consistency of tissue dimensions while maintaining cell viability. Printing with 30 G tips resulted in decreased mean wall thickness (318.6<i>μ</i>m) without compromising mean cell viability (94.8%). Histological analysis of cell-only smooth muscle tissues cultured for 14 d in OMA support baths exhibited decreased wall thickness using 30 G dispensing tips, which correlated with increased collagen deposition and alignment. In addition, a TUNEL assay indicated a decrease in cell death in tissues printed with thinner (30 G) dispensing tips. Mechanical testing demonstrated that tissues printed with a 30 G dispensing tip exhibit an increase in ultimate tensile strength compared to those printed with a 27 G dispensing tip. Overall, these findings highlight the importance of precise control over bioprinting parameters to generate mechanically robust tissues when using cell-only bioinks dispensed and cultured within hydrogel support baths. The ability to control print dimensions using cell-only bioinks may enable bioprinting of more complex soft tissue geometries to generate<i>in vitro</i>tissue models.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911585","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring bio-nanomaterials as antibiotic allies to combat antimicrobial resistance. 探索作为抗生素盟友的生物纳米材料,以对抗抗菌药耐药性。
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-08-29 DOI: 10.1088/1758-5090/ad6b45
Bindiya Barsola, Shivani Saklani, Diksha Pathania, Priyanka Kumari, Sonu Sonu, Sarvesh Rustagi, Pardeep Singh, Pankaj Raizada, Tae Seok Moon, Ajeet Kaushik, Vishal Chaudhary

Antimicrobial resistance (AMR) poses an emergent threat to global health due to antibiotic abuse, overuse and misuse, necessitating urgent innovative and sustainable solutions. The utilization of bio-nanomaterials as antibiotic allies is a green, economic, sustainable and renewable strategy to combat this pressing issue. These biomaterials involve green precursors (e.g. biowaste, plant extracts, essential oil, microbes, and agricultural residue) and techniques for their fabrication, which reduce their cyto/environmental toxicity and exhibit economic manufacturing, enabling a waste-to-wealth circular economy module. Their nanoscale dimensions with augmented biocompatibility characterize bio-nanomaterials and offer distinctive advantages in addressing AMR. Their ability to target pathogens, such as bacteria and viruses, at the molecular level, coupled with their diverse functionalities and bio-functionality doping from natural precursors, allows for a multifaceted approach to combat resistance. Furthermore, bio-nanomaterials can be tailored to enhance the efficacy of existing antimicrobial agents or deliver novel therapies, presenting a versatile platform for innovation. Their use in combination with traditional antibiotics can mitigate resistance mechanisms, prolong the effectiveness of existing treatments, and reduce side effects. This review aims to shed light on the potential of bio-nanomaterials in countering AMR, related mechanisms, and their applications in various domains. These roles encompass co-therapy, nanoencapsulation, and antimicrobial stewardship, each offering a distinct avenue for overcoming AMR. Besides, it addresses the challenges associated with bio-nanomaterials, emphasizing the importance of regulatory considerations. These green biomaterials are the near future of One Health Care, which will have economic, non-polluting, non-toxic, anti-resistant, biocompatible, degradable, and repurposable avenues, contributing to sustainable development goals.

由于抗生素的滥用、过度使用和误用,抗菌素耐药性(AMR)对全球健康构成了新的威胁,因此迫切需要创新和可持续的解决方案。利用生物纳米材料作为抗生素盟友,是应对这一紧迫问题的绿色、经济、可持续和可再生战略。这些生物材料涉及绿色前体(如生物废料、植物提取物、精油、微生物和农业残留物)及其制造技术,可降低其细胞/环境毒性,并表现出经济的制造方式,从而实现从废物到财富的循环经济模块。生物纳米材料具有纳米级尺寸和更强的生物相容性,在应对 AMR 方面具有独特的优势。生物纳米材料能够在分子水平上靶向细菌和病毒等病原体,再加上其多样化的功能性以及从天然前体中掺入的生物功能性,使得生物纳米材料能够以多层面的方式对抗抗药性。此外,生物纳米材料可以量身定制,以增强现有抗菌剂的功效或提供新型疗法,从而为创新提供了一个多功能平台。将生物纳米材料与传统抗生素结合使用,可减轻抗药性机制,延长现有疗法的疗效,并减少副作用。本综述旨在阐明生物纳米材料在对抗 AMR 方面的潜力、相关机制及其在各个领域的应用。这些作用包括共同治疗、纳米封装和抗菌管理,每种作用都为克服 AMR 提供了独特的途径。此外,该书还探讨了与生物纳米材料相关的挑战,强调了监管考虑因素的重要性。这些绿色生物材料是 "一种医疗保健 "的不远的将来,它们将具有经济、无污染、无毒、抗抗性、生物相容性、可降解和可再利用的途径,有助于实现可持续发展目标。
{"title":"Exploring bio-nanomaterials as antibiotic allies to combat antimicrobial resistance.","authors":"Bindiya Barsola, Shivani Saklani, Diksha Pathania, Priyanka Kumari, Sonu Sonu, Sarvesh Rustagi, Pardeep Singh, Pankaj Raizada, Tae Seok Moon, Ajeet Kaushik, Vishal Chaudhary","doi":"10.1088/1758-5090/ad6b45","DOIUrl":"10.1088/1758-5090/ad6b45","url":null,"abstract":"<p><p>Antimicrobial resistance (AMR) poses an emergent threat to global health due to antibiotic abuse, overuse and misuse, necessitating urgent innovative and sustainable solutions. The utilization of bio-nanomaterials as antibiotic allies is a green, economic, sustainable and renewable strategy to combat this pressing issue. These biomaterials involve green precursors (e.g. biowaste, plant extracts, essential oil, microbes, and agricultural residue) and techniques for their fabrication, which reduce their cyto/environmental toxicity and exhibit economic manufacturing, enabling a waste-to-wealth circular economy module. Their nanoscale dimensions with augmented biocompatibility characterize bio-nanomaterials and offer distinctive advantages in addressing AMR. Their ability to target pathogens, such as bacteria and viruses, at the molecular level, coupled with their diverse functionalities and bio-functionality doping from natural precursors, allows for a multifaceted approach to combat resistance. Furthermore, bio-nanomaterials can be tailored to enhance the efficacy of existing antimicrobial agents or deliver novel therapies, presenting a versatile platform for innovation. Their use in combination with traditional antibiotics can mitigate resistance mechanisms, prolong the effectiveness of existing treatments, and reduce side effects. This review aims to shed light on the potential of bio-nanomaterials in countering AMR, related mechanisms, and their applications in various domains. These roles encompass co-therapy, nanoencapsulation, and antimicrobial stewardship, each offering a distinct avenue for overcoming AMR. Besides, it addresses the challenges associated with bio-nanomaterials, emphasizing the importance of regulatory considerations. These green biomaterials are the near future of One Health Care, which will have economic, non-polluting, non-toxic, anti-resistant, biocompatible, degradable, and repurposable avenues, contributing to sustainable development goals.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892725","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Highly conductive, stretchable, and biocompatible graphene oxide biocomposite hydrogel for advanced tissue engineering. 用于先进组织工程的高导电性、可拉伸和生物相容性氧化石墨烯生物复合水凝胶。
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-08-28 DOI: 10.1088/1758-5090/ad6cf7
Young Jin Lee, Olatunji Ajiteru, Ji Seung Lee, Ok Joo Lee, Kyu Young Choi, Soon Hee Kim, Chan Hum Park

The importance of hydrogels in tissue engineering cannot be overemphasized due to their resemblance to the native extracellular matrix. However, natural hydrogels with satisfactory biocompatibility exhibit poor mechanical behavior, which hampers their application in stress-bearing soft tissue engineering. Here, we describe the fabrication of a double methacrylated gelatin bioink covalently linked to graphene oxide (GO) via a zero-length crosslinker, digitally light-processed (DLP) printable into 3D complex structures with high fidelity. The resultant natural hydrogel (GelGOMA) exhibits a conductivity of 15.0 S m-1as a result of the delocalization of theπ-orbital from the covalently linked GO. Furthermore, the hydrogel shows a compressive strength of 1.6 MPa, and a 2.0 mm thick GelGOMA can withstand a 1.0 kg ms-1momentum. The printability and mechanical strengths of GelGOMAs were demonstrated by printing a fish heart with a functional fluid pumping mechanism and tricuspid valves. Its biocompatibility, electroconductivity, and physiological relevance enhanced the proliferation and differentiation of myoblasts and neuroblasts and the contraction of human-induced pluripotent stem cell-derived cardiomyocytes. GelGOMA demonstrates the potential for the tissue engineering of functional hearts and wearable electronic devices.

水凝胶与原生细胞外基质(ECM)相似,因此在组织工程中的重要性怎么强调都不为过。然而,生物相容性令人满意的天然水凝胶却表现出很差的机械性能,这阻碍了它们在承压软组织工程中的应用。在这里,我们介绍了通过零长交联剂与氧化石墨烯(GO)共价连接的双甲基丙烯酸明胶生物墨水的制备方法,该方法可通过数字光处理(DLP)打印成高保真的三维复杂结构。由于共价连接的 GO 的 π 轨道发生了偏移,因此生成的天然水凝胶(GelGOMA)的电导率达到了 15.0 S m-1。此外,这种水凝胶的抗压强度为 1.6 兆帕,2.0 毫米厚的 GelGOMA 可以承受 1.0 千克毫秒-1 的动量。通过打印具有功能性液体泵送机制和三尖瓣的鱼心脏,证明了 GelGOMA 的可打印性和机械性能。它的生物相容性、导电性和生理相关性增强了成肌细胞和神经母细胞的增殖和分化,以及源自 hiPSC 的心肌细胞的收缩。GelGOMA展示了功能性心脏组织工程和可穿戴电子设备的潜力。
{"title":"Highly conductive, stretchable, and biocompatible graphene oxide biocomposite hydrogel for advanced tissue engineering.","authors":"Young Jin Lee, Olatunji Ajiteru, Ji Seung Lee, Ok Joo Lee, Kyu Young Choi, Soon Hee Kim, Chan Hum Park","doi":"10.1088/1758-5090/ad6cf7","DOIUrl":"10.1088/1758-5090/ad6cf7","url":null,"abstract":"<p><p>The importance of hydrogels in tissue engineering cannot be overemphasized due to their resemblance to the native extracellular matrix. However, natural hydrogels with satisfactory biocompatibility exhibit poor mechanical behavior, which hampers their application in stress-bearing soft tissue engineering. Here, we describe the fabrication of a double methacrylated gelatin bioink covalently linked to graphene oxide (GO) via a zero-length crosslinker, digitally light-processed (DLP) printable into 3D complex structures with high fidelity. The resultant natural hydrogel (GelGOMA) exhibits a conductivity of 15.0 S m<sup>-1</sup>as a result of the delocalization of the<i>π</i>-orbital from the covalently linked GO. Furthermore, the hydrogel shows a compressive strength of 1.6 MPa, and a 2.0 mm thick GelGOMA can withstand a 1.0 kg ms<sup>-1</sup>momentum. The printability and mechanical strengths of GelGOMAs were demonstrated by printing a fish heart with a functional fluid pumping mechanism and tricuspid valves. Its biocompatibility, electroconductivity, and physiological relevance enhanced the proliferation and differentiation of myoblasts and neuroblasts and the contraction of human-induced pluripotent stem cell-derived cardiomyocytes. GelGOMA demonstrates the potential for the tissue engineering of functional hearts and wearable electronic devices.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141905827","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Constraint based Bayesian optimization of bioink precursor: a machine learning framework. 基于约束的生物墨水前体贝叶斯优化:一种机器学习框架。
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-08-28 DOI: 10.1088/1758-5090/ad716e
Yihao Xu, Rokeya Sarah, Ahasan Habib, Yongmin Liu, Bashir Khoda

Current research practice for optimizing bioink involves exhaustive experimentation with multi-material composition for determining the printability, shape fidelity and biocompatibility. Predicting bioink properties can be beneficial to the research community but is a challenging task due to the non-Newtonian behavior in complex composition. Existing models such as Cross model become inadequate for predicting the viscosity for heterogeneous composition of bioinks. In this paper, we utilize a machine learning framework to accurately predict the viscosity of heterogeneous bioink compositions, aiming to enhance extrusion-based bioprinting techniques. Utilizing Bayesian optimization (BO), our strategy leverages a limited dataset to inform our model. This is a technique especially useful of the typically sparse data in this domain. Moreover, we have also developed a mask technique that can handle complex constraints, informed by domain expertise, to define the feasible parameter space for the components of the bioink and their interactions. Our proposed method is focused on predicting the intrinsic factor (e.g. viscosity) of the bioink precursor which is tied to the extrinsic property (e.g. cell viability) through the mask function. Through the optimization of the hyperparameter, we strike a balance between exploration of new possibilities and exploitation of known data, a balance crucial for refining our acquisition function. This function then guides the selection of subsequent sampling points within the defined viable space and the process continues until convergence is achieved, indicating that the model has sufficiently explored the parameter space and identified the optimal or near-optimal solutions. Employing this AI-guided BO framework, we have developed, tested, and validated a surrogate model for determining the viscosity of heterogeneous bioink compositions. This data-driven approach significantly reduces the experimental workload required to identify bioink compositions conducive to functional tissue growth. It not only streamlines the process of finding the optimal bioink compositions from a vast array of heterogeneous options but also offers a promising avenue for accelerating advancements in tissue engineering by minimizing the need for extensive experimental trials.

目前,优化生物墨水的研究实践包括对多种材料成分进行详尽的实验,以确定其可印刷性、形状保真度和生物相容性。预测生物墨水的特性对研究界大有裨益,但由于复杂成分中的非牛顿行为,预测生物墨水的特性是一项极具挑战性的任务。现有模型(如 Cross 模型)不足以预测生物墨水异质成分的粘度。在本文中,我们利用机器学习框架来准确预测异质生物墨水成分的粘度,旨在提高基于挤压的生物打印技术。利用贝叶斯优化(BO),我们的策略是利用有限的数据集为我们的模型提供信息。这种技术对该领域典型的稀疏数据尤为有用。此外,我们还开发了一种掩膜技术,可以处理复杂的约束条件,并通过领域专业知识来定义生物墨水成分及其相互作用的可行参数空间。我们提出的方法侧重于预测生物墨水前体的内在因素(如粘度),通过掩膜函数将其与外在特性(如细胞活力)联系起来。通过优化超参数,我们在探索新的可能性和利用已知数据之间取得了平衡,这种平衡对于完善我们的采集功能至关重要。然后,该函数将指导在已定义的可行空间内选择后续采样点,这一过程一直持续到收敛为止,表明模型已充分探索了参数空间,并确定了最优或接近最优的解决方案。利用这种人工智能指导的 BO 框架,我们开发、测试并验证了一种用于确定异质生物墨水成分粘度的替代模型。这种数据驱动方法大大减少了确定有利于功能性组织生长的生物墨水成分所需的实验工作量。它不仅简化了从大量异质选择中寻找最佳生物墨水成分的过程,而且通过最大限度地减少对大量实验的需求,为加快组织工程的发展提供了一条大有可为的途径。
{"title":"Constraint based Bayesian optimization of bioink precursor: a machine learning framework.","authors":"Yihao Xu, Rokeya Sarah, Ahasan Habib, Yongmin Liu, Bashir Khoda","doi":"10.1088/1758-5090/ad716e","DOIUrl":"10.1088/1758-5090/ad716e","url":null,"abstract":"<p><p>Current research practice for optimizing bioink involves exhaustive experimentation with multi-material composition for determining the printability, shape fidelity and biocompatibility. Predicting bioink properties can be beneficial to the research community but is a challenging task due to the non-Newtonian behavior in complex composition. Existing models such as Cross model become inadequate for predicting the viscosity for heterogeneous composition of bioinks. In this paper, we utilize a machine learning framework to accurately predict the viscosity of heterogeneous bioink compositions, aiming to enhance extrusion-based bioprinting techniques. Utilizing Bayesian optimization (BO), our strategy leverages a limited dataset to inform our model. This is a technique especially useful of the typically sparse data in this domain. Moreover, we have also developed a mask technique that can handle complex constraints, informed by domain expertise, to define the feasible parameter space for the components of the bioink and their interactions. Our proposed method is focused on predicting the intrinsic factor (e.g. viscosity) of the bioink precursor which is tied to the extrinsic property (e.g. cell viability) through the mask function. Through the optimization of the hyperparameter, we strike a balance between exploration of new possibilities and exploitation of known data, a balance crucial for refining our acquisition function. This function then guides the selection of subsequent sampling points within the defined viable space and the process continues until convergence is achieved, indicating that the model has sufficiently explored the parameter space and identified the optimal or near-optimal solutions. Employing this AI-guided BO framework, we have developed, tested, and validated a surrogate model for determining the viscosity of heterogeneous bioink compositions. This data-driven approach significantly reduces the experimental workload required to identify bioink compositions conducive to functional tissue growth. It not only streamlines the process of finding the optimal bioink compositions from a vast array of heterogeneous options but also offers a promising avenue for accelerating advancements in tissue engineering by minimizing the need for extensive experimental trials.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142008232","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Complexin vitromodels positioned for impact to drug testing in pharma: a review. 络氨酸维生素模型对制药业药物测试的影响:综述。
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-08-27 DOI: 10.1088/1758-5090/ad6933
Serah Kang, Eugene C Chen, Helen Cifuentes, Julia Y Co, Gabrielle Cole, Jessica Graham, Rebecca Hsia, Tomomi Kiyota, Jessica A Klein, Katharina T Kroll, Lenitza M Nieves Lopez, Leah M Norona, Heshan Peiris, Ratnakar Potla, Monica Romero-Lopez, Julien G Roth, Min Tseng, Aaron M Fullerton, Kimberly A Homan

Recent years have seen the creation and popularization of various complexin vitromodels (CIVMs), such as organoids and organs-on-chip, as a technology with the potential to reduce animal usage in pharma while also enhancing our ability to create safe and efficacious drugs for patients. Public awareness of CIVMs has increased, in part, due to the recent passage of the FDA Modernization Act 2.0. This visibility is expected to spur deeper investment in and adoption of such models. Thus, end-users and model developers alike require a framework to both understand the readiness of current models to enter the drug development process, and to assess upcoming models for the same. This review presents such a framework for model selection based on comparative -omics data (which we term model-omics), and metrics for qualification of specific test assays that a model may support that we term context-of-use (COU) assays. We surveyed existing healthy tissue models and assays for ten drug development-critical organs of the body, and provide evaluations of readiness and suggestions for improving model-omics and COU assays for each. In whole, this review comes from a pharma perspective, and seeks to provide an evaluation of where CIVMs are poised for maximum impact in the drug development process, and a roadmap for realizing that potential.

近年来,有机体和片上器官等各种复合蛋白体模(CIVMs)作为一种有可能减少制药中动物用量的技术不断涌现和普及,同时也提高了我们为患者研制安全有效药物的能力。公众对 CIVMs 的认识有所提高,部分原因是最近通过了《美国食品及药物管理局现代化法案 2.0》。这种知名度有望促进对此类模型的更多投资和采用。因此,终端用户和模型开发者都需要一个框架,既能了解当前模型进入药物开发流程的准备情况,又能对即将推出的模型进行评估。本综述提出了这样一个框架:基于比较组学数据(我们称之为模型组学)选择模型,并对模型可能支持的特定测试化验进行鉴定,我们称之为使用情境(COU)化验。我们调查了人体十个药物开发关键器官的现有健康组织模型和检测方法,并对每个器官的模型组学和 COU 检测方法的准备情况进行了评估,并提出了改进建议。总之,本综述从制药业的角度出发,旨在评估 CIVM 在药物开发过程中可发挥最大影响的领域,并提供实现这一潜力的路线图。
{"title":"Complex<i>in vitro</i>models positioned for impact to drug testing in pharma: a review.","authors":"Serah Kang, Eugene C Chen, Helen Cifuentes, Julia Y Co, Gabrielle Cole, Jessica Graham, Rebecca Hsia, Tomomi Kiyota, Jessica A Klein, Katharina T Kroll, Lenitza M Nieves Lopez, Leah M Norona, Heshan Peiris, Ratnakar Potla, Monica Romero-Lopez, Julien G Roth, Min Tseng, Aaron M Fullerton, Kimberly A Homan","doi":"10.1088/1758-5090/ad6933","DOIUrl":"10.1088/1758-5090/ad6933","url":null,"abstract":"<p><p>Recent years have seen the creation and popularization of various complex<i>in vitro</i>models (CIVMs), such as organoids and organs-on-chip, as a technology with the potential to reduce animal usage in pharma while also enhancing our ability to create safe and efficacious drugs for patients. Public awareness of CIVMs has increased, in part, due to the recent passage of the FDA Modernization Act 2.0. This visibility is expected to spur deeper investment in and adoption of such models. Thus, end-users and model developers alike require a framework to both understand the readiness of current models to enter the drug development process, and to assess upcoming models for the same. This review presents such a framework for model selection based on comparative -omics data (which we term model-omics), and metrics for qualification of specific test assays that a model may support that we term context-of-use (COU) assays. We surveyed existing healthy tissue models and assays for ten drug development-critical organs of the body, and provide evaluations of readiness and suggestions for improving model-omics and COU assays for each. In whole, this review comes from a pharma perspective, and seeks to provide an evaluation of where CIVMs are poised for maximum impact in the drug development process, and a roadmap for realizing that potential.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142071942","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Microneedle system for tissue engineering and regenerative medicines: a smart and efficient therapeutic approach. 用于组织工程和再生医学的微针系统:一种智能高效的治疗方法。
IF 8.2 2区 医学 Q1 ENGINEERING, BIOMEDICAL Pub Date : 2024-08-27 DOI: 10.1088/1758-5090/ad6d90
Muhammad Umar Aslam Khan, Muhammad Azhar Aslam, Mohd Faizal Bin Abdullah, Hilal Gul, Goran M Stojanović, Abdalla Abdal-Hay, Anwarul Hasan

The global demand for an enhanced quality of life and extended lifespan has driven significant advancements in tissue engineering and regenerative medicine. These fields utilize a range of interdisciplinary theories and techniques to repair structurally impaired or damaged tissues and organs, as well as restore their normal functions. Nevertheless, the clinical efficacy of medications, materials, and potent cells used at the laboratory level is always constrained by technological limitations. A novel platform known as adaptable microneedles has been developed to address the abovementioned issues. These microneedles offer a solution for the localized distribution of various cargos while minimizing invasiveness. Microneedles provide favorable patient compliance in clinical settings due to their effective administration and ability to provide a painless and convenient process. In this review article, we summarized the most recent development of microneedles, and we started by classifying various microneedle systems, advantages, and fundamental properties. Subsequently, it provides a comprehensive overview of different types of microneedles, the material used to fabricate microneedles, the fundamental properties of ideal microneedles, and their applications in tissue engineering and regenerative medicine, primarily focusing on preserving and restoring impaired tissues and organs. The limitations and perspectives have been discussed by concluding their future therapeutic applications in tissue engineering and regenerative medicines.

全球对提高生活质量和延长寿命的需求推动了组织工程和再生医学的长足发展。这些领域利用一系列跨学科理论和技术来修复结构受损或损坏的组织和器官,并恢复其正常功能。然而,在实验室层面使用的药物、材料和强效细胞的临床疗效始终受到技术限制。为了解决上述问题,我们开发了一种称为可适应微针的新型平台。这些微针为各种载体的局部分布提供了解决方案,同时最大限度地减少了侵入性。由于微针能有效给药,且能提供无痛、方便的治疗过程,因此在临床环境中为患者提供了良好的依从性。在这篇综述文章中,我们总结了微针的最新发展,首先对各种微针系统、优势和基本特性进行了分类。随后,文章全面概述了不同类型的微针、用于制造微针的材料、理想微针的基本特性,以及它们在组织工程和再生医学中的应用,主要侧重于保存和恢复受损的组织和器官。通过总结微针在组织工程和再生医学中的未来治疗应用,讨论了其局限性和前景。
{"title":"Microneedle system for tissue engineering and regenerative medicines: a smart and efficient therapeutic approach.","authors":"Muhammad Umar Aslam Khan, Muhammad Azhar Aslam, Mohd Faizal Bin Abdullah, Hilal Gul, Goran M Stojanović, Abdalla Abdal-Hay, Anwarul Hasan","doi":"10.1088/1758-5090/ad6d90","DOIUrl":"10.1088/1758-5090/ad6d90","url":null,"abstract":"<p><p>The global demand for an enhanced quality of life and extended lifespan has driven significant advancements in tissue engineering and regenerative medicine. These fields utilize a range of interdisciplinary theories and techniques to repair structurally impaired or damaged tissues and organs, as well as restore their normal functions. Nevertheless, the clinical efficacy of medications, materials, and potent cells used at the laboratory level is always constrained by technological limitations. A novel platform known as adaptable microneedles has been developed to address the abovementioned issues. These microneedles offer a solution for the localized distribution of various cargos while minimizing invasiveness. Microneedles provide favorable patient compliance in clinical settings due to their effective administration and ability to provide a painless and convenient process. In this review article, we summarized the most recent development of microneedles, and we started by classifying various microneedle systems, advantages, and fundamental properties. Subsequently, it provides a comprehensive overview of different types of microneedles, the material used to fabricate microneedles, the fundamental properties of ideal microneedles, and their applications in tissue engineering and regenerative medicine, primarily focusing on preserving and restoring impaired tissues and organs. The limitations and perspectives have been discussed by concluding their future therapeutic applications in tissue engineering and regenerative medicines.</p>","PeriodicalId":8964,"journal":{"name":"Biofabrication","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911583","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Biofabrication
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