1
|
Gopallawa I, Gupta C, Jawa R, Cyril A, Jawa V, Chirmule N, Gujar V. Applications of Organoids in Advancing Drug Discovery and Development. J Pharm Sci 2024; 113:2659-2667. [PMID: 39002723 DOI: 10.1016/j.xphs.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/15/2024]
Abstract
Organoids are small, self-organizing three-dimensional cell cultures that are derived from stem cells or primary organs. These cultures replicate the complexity of an organ, which cannot be achieved by single-cell culture systems. Organoids can be used in testing of new drugs instead of animals. Development and validation of organoids is thus important to reduce the reliance on animals for drug testing. In this review, we have discussed the developmental and regulatory aspects of organoids and highlighted their importance in drug development. We have first summarized different types of culture-based organoid systems such as submerged Matrigel, micro-fluidic 3D cultures, inducible pluripotent stem cells, and air-liquid interface cultures. These systems help us understand the intricate interplay between cells and their surrounding milieu for identifying functions of target receptors, soluble factors, and spatial interactions. Further, we have discussed the advances in humanized severe-combined immunodeficiency mouse models and their applications in the pharmacology of immune-oncology. Since regulatory aspects are important in using organoids for drug development, we have summarized FDA and EMA regulations on organoid research to support pre-clinical studies. Finally, we have included some unique studies highlighting the use of organoids in studying infectious diseases, cancer, and fundamental biology. These studies also exemplify the latest technological advances in organoid development resulting in improved efficiency. Overall, this review comprehensively summarizes the applications of organoids in early drug development during discovery and pre-clinical studies.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | | | - Rayan Jawa
- University of Pennsylvania, Philadelphia, PA, USA
| | - Arya Cyril
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Vibha Jawa
- Bristol Myers Squibb, Lawrenceville, NY, USA.
| | | | - Vikramsingh Gujar
- Anatomy and Cell Biology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| |
Collapse
|
2
|
Thaden O, Schneider N, Walther T, Spiller E, Taoum A, Göpfrich K, Duarte Campos D. Bioprinting of Synthetic Cell-like Lipid Vesicles to Augment the Functionality of Tissues after Manufacturing. ACS Synth Biol 2024; 13:2436-2446. [PMID: 39025476 PMCID: PMC11334175 DOI: 10.1021/acssynbio.4c00137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024]
Abstract
Bioprinting is an automated bioassembly method that enables the formation of human tissue-like constructs to restore or replace damaged tissues. Regardless of the employed bioprinting method, cells undergo mechanical stress that can impact their survival and function postprinting. In this study, we investigate the use of a synthetic cell-like unit, giant unilamellar vesicles (GUVs), as adjuvants of the cellular function of human cells postprinting, or in future as the complete replacement of human cells. We analyzed the impact of two nozzle-based bioprinting methods (drop-on-demand and extrusion bioprinting) on the structure, stability, and function of GUVs. We showed that over 65% of the GUVs remain intact when printing at 0.5 bar, demonstrating the potential of using GUVs as a synthetic cell source. We further increased the stability of GUVs in a cell culture medium by introducing polyethylene glycol (PEG) into the GUV lipid membrane. The presence of PEG, however, diminished the structural properties of GUVs postprinting, and reduced the interaction of GUVs with human cells. Although the design of PEG-GUVs can still be modified in future studies for better cell-GUV interactions, we demonstrated that GUVs are functional postprinting. Chlorin e6-PEG-GUVs loaded with a fluorescent dye were bioprinted, and they released the dye postprinting only upon illumination. This is a new strategy to deliver carriers, such as growth factors, drugs, nutrients, or gases, inside large bioprinted specimens on a millimeter to centimeter scale. Overall, we showed that printed GUVs can augment the functionality of manufactured human tissues.
Collapse
Affiliation(s)
- Ole Thaden
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| | - Nicole Schneider
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| | - Tobias Walther
- Biophysical
Engineering of Life Group, Center for Molecular
Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
- Max
Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Erin Spiller
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| | - Alexandre Taoum
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| | - Kerstin Göpfrich
- Biophysical
Engineering of Life Group, Center for Molecular
Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
- Max
Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Daniela Duarte Campos
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| |
Collapse
|
3
|
Wang X, Chen Q, Li J, Tian W, Liu Z, Chen T. Recent adavances of functional modules for tooth regeneration. J Mater Chem B 2024; 12:7497-7518. [PMID: 39021127 DOI: 10.1039/d4tb01027b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Dental diseases, such as dental caries and periodontal disorders, constitute a major global health challenge, affecting millions worldwide and often resulting in tooth loss. Traditional dental treatments, though beneficial, typically cannot fully restore the natural functions and structures of teeth. This limitation has prompted growing interest in innovative strategies for tooth regeneration methods. Among these, the use of dental stem cells to generate functional tooth modules represents an emerging and promising approach in dental tissue engineering. These modules aim to closely replicate the intricate morphology and essential physiological functions of dental tissues. Recent advancements in regenerative research have not only enhanced the assembly techniques for these modules but also highlighted their therapeutic potential in addressing various dental diseases. In this review, we discuss the latest progress in the construction of functional tooth modules, especially on regenerating dental pulp, periodontal tissue, and tooth roots.
Collapse
Affiliation(s)
- Xuan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Qiuyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jiayi Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhi Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Tian Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
4
|
Kim W, Kim G. Engineered 3D liver-tissue model with minispheroids formed by a bioprinting process supported with in situ electrical stimulation. Bioact Mater 2024; 35:382-400. [PMID: 38379698 PMCID: PMC10876469 DOI: 10.1016/j.bioactmat.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/22/2024] Open
Abstract
Three-dimensional (3D) bioprinting, an effective technique for building cell-laden structures providing native extracellular matrix environments, presents challenges, including inadequate cellular interactions. To address these issues, cell spheroids offer a promising solution for improving their biological functions. Particularly, minispheroids with 50-100 μm diameters exhibit enhanced cellular maturation. We propose a one-step minispheroid-forming bioprinting process incorporating electrical stimulation (E-MS-printing). By stimulating the cells, minispheroids with controlled diameters were generated by manipulating the bioink viscosity and stimulation intensity. To validate its feasibility, E-MS-printing process was applied to fabricate an engineered liver model designed to mimic the hepatic lobule unit. E-MS-printing was employed to print the hepatocyte region, followed by bioprinting the central vein using a core-shell nozzle. The resulting constructs displayed native liver-mimetic structures containing minispheroids, which facilitated improved hepatic cell maturation, functional attributes, and vessel formation. Our results demonstrate a new potential 3D liver model that can replicate native liver tissues.
Collapse
Affiliation(s)
- WonJin Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
| | - GeunHyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
5
|
Puertas-Bartolomé M, Venegas-Bustos D, Acosta S, Rodríguez-Cabello JC. Contribution of the ELRs to the development of advanced in vitro models. Front Bioeng Biotechnol 2024; 12:1363865. [PMID: 38650751 PMCID: PMC11033926 DOI: 10.3389/fbioe.2024.1363865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Developing in vitro models that accurately mimic the microenvironment of biological structures or processes holds substantial promise for gaining insights into specific biological functions. In the field of tissue engineering and regenerative medicine, in vitro models able to capture the precise structural, topographical, and functional complexity of living tissues, prove to be valuable tools for comprehending disease mechanisms, assessing drug responses, and serving as alternatives or complements to animal testing. The choice of the right biomaterial and fabrication technique for the development of these in vitro models plays an important role in their functionality. In this sense, elastin-like recombinamers (ELRs) have emerged as an important tool for the fabrication of in vitro models overcoming the challenges encountered in natural and synthetic materials due to their intrinsic properties, such as phase transition behavior, tunable biological properties, viscoelasticity, and easy processability. In this review article, we will delve into the use of ELRs for molecular models of intrinsically disordered proteins (IDPs), as well as for the development of in vitro 3D models for regenerative medicine. The easy processability of the ELRs and their rational design has allowed their use for the development of spheroids and organoids, or bioinks for 3D bioprinting. Thus, incorporating ELRs into the toolkit of biomaterials used for the fabrication of in vitro models, represents a transformative step forward in improving the accuracy, efficiency, and functionality of these models, and opening up a wide range of possibilities in combination with advanced biofabrication techniques that remains to be explored.
Collapse
Affiliation(s)
- María Puertas-Bartolomé
- Technical Proteins Nanobiotechnology, S.L. (TPNBT), Valladolid, Spain
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Desiré Venegas-Bustos
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Sergio Acosta
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
6
|
Dranseike D, Ota Y, Edwardson TGW, Guzzi EA, Hori M, Nakic ZR, Deshmukh DV, Levasseur MD, Mattli K, Tringides CM, Zhou J, Hilvert D, Peters C, Tibbitt MW. Designed modular protein hydrogels for biofabrication. Acta Biomater 2024; 177:107-117. [PMID: 38382830 DOI: 10.1016/j.actbio.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
Designing proteins that fold and assemble over different length scales provides a way to tailor the mechanical properties and biological performance of hydrogels. In this study, we designed modular proteins that self-assemble into fibrillar networks and, as a result, form hydrogel materials with novel properties. We incorporated distinct functionalities by connecting separate self-assembling (A block) and cell-binding (B block) domains into single macromolecules. The number of self-assembling domains affects the rigidity of the fibers and the final storage modulus G' of the materials. The mechanical properties of the hydrogels could be tuned over a broad range (G' = 0.1 - 10 kPa), making them suitable for the cultivation and differentiation of multiple cell types, including cortical neurons and human mesenchymal stem cells. Moreover, we confirmed the bioavailability of cell attachment domains in the hydrogels that can be further tailored for specific cell types or other biological applications. Finally, we demonstrate the versatility of the designed proteins for application in biofabrication as 3D scaffolds that support cell growth and guide their function. STATEMENT OF SIGNIFICANCE: Designed proteins that enable the decoupling of biophysical and biochemical properties within the final material could enable modular biomaterial engineering. In this context, we present a designed modular protein platform that integrates self-assembling domains (A blocks) and cell-binding domains (B blocks) within a single biopolymer. The linking of assembly domains and cell-binding domains this way provided independent tuning of mechanical properties and inclusion of biofunctional domains. We demonstrate the use of this platform for biofabrication, including neural cell culture and 3D printing of scaffolds for mesenchymal stem cell culture and differentiation. Overall, this work highlights how informed design of biopolymer sequences can enable the modular design of protein-based hydrogels with independently tunable biophysical and biochemical properties.
Collapse
Affiliation(s)
- Dalia Dranseike
- Macromolecular Engineering Laboratory, ETH Zurich, Zurich, Switzerland
| | - Yusuke Ota
- Organic Chemistry Laboratory, ETH Zurich, Zurich, Switzerland
| | | | - Elia A Guzzi
- Macromolecular Engineering Laboratory, ETH Zurich, Zurich, Switzerland
| | - Mao Hori
- Organic Chemistry Laboratory, ETH Zurich, Zurich, Switzerland
| | | | | | | | - Kevin Mattli
- Biosystems Technology, ZHAW, Wädenswil, Switzerland
| | | | - Jiangtao Zhou
- Laboratory of Food and Soft Materials, ETH Zurich, Switzerland
| | - Donald Hilvert
- Organic Chemistry Laboratory, ETH Zurich, Zurich, Switzerland.
| | | | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
7
|
Koskinen LM, Nieminen L, Arjonen A, Guzmán C, Peurla M, Peuhu E. Spatial Engineering of Mammary Epithelial Cell Cultures with 3D Bioprinting Reveals Growth Control by Branch Point Proximity. J Mammary Gland Biol Neoplasia 2024; 29:5. [PMID: 38416267 PMCID: PMC10902034 DOI: 10.1007/s10911-024-09557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024] Open
Abstract
The three-dimensional (3D) structure of the ductal epithelium and the surrounding extracellular matrix (ECM) are integral aspects of the breast tissue, and they have important roles during mammary gland development, function and malignancy. However, the architecture of the branched mammary epithelial network is poorly recapitulated in the current in vitro models. 3D bioprinting is an emerging approach to improve tissue-mimicry in cell culture. Here, we developed and optimized a protocol for 3D bioprinting of normal and cancerous mammary epithelial cells into a branched Y-shape to study the role of cell positioning in the regulation of cell proliferation and invasion. Non-cancerous cells formed continuous 3D cell networks with several organotypic features, whereas the ductal carcinoma in situ (DCIS) -like cancer cells exhibited aberrant basal polarization and defective formation of the basement membrane (BM). Quantitative analysis over time demonstrated that both normal and cancerous cells proliferate more at the branch tips compared to the trunk region of the 3D-bioprinted cultures, and particularly at the tip further away from the branch point. The location-specific rate of proliferation was independent of TGFβ signaling but invasion of the DCIS-like breast cancer cells was reduced upon the inhibition of TGFβ. Thus, our data demonstrate that the 3D-bioprinted cells can sense their position in the branched network of cells and proliferate at the tips, thus recapitulating this feature of mammary epithelial branching morphogenesis. In all, our results demonstrate the capacity of the developed 3D bioprinting method for quantitative analysis of the relationships between tissue structure and cell behavior in breast morphogenesis and cancer.
Collapse
Affiliation(s)
- Leena M Koskinen
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | | | | | - Markus Peurla
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Emilia Peuhu
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| |
Collapse
|
8
|
Arjoca S, Bojin F, Neagu M, Păunescu A, Neagu A, Păunescu V. Hydrogel Extrusion Speed Measurements for the Optimization of Bioprinting Parameters. Gels 2024; 10:103. [PMID: 38391433 PMCID: PMC10888060 DOI: 10.3390/gels10020103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Three-dimensional (3D) bioprinting is the use of computer-controlled transfer processes for assembling bioinks (cell clusters or materials loaded with cells) into structures of prescribed 3D organization. The correct bioprinting parameters ensure a fast and accurate bioink deposition without exposing the cells to harsh conditions. This study seeks to optimize pneumatic extrusion-based bioprinting based on hydrogel flow rate and extrusion speed measurements. We measured the rate of the hydrogel flow through a cylindrical nozzle and used non-Newtonian hydrodynamics to fit the results. From the videos of free-hanging hydrogel strands delivered from a stationary print head, we inferred the extrusion speed, defined as the speed of advancement of newly formed strands. Then, we relied on volume conservation to evaluate the extrudate swell ratio. The theoretical analysis enabled us to compute the extrusion speed for pressures not tested experimentally as well as the printing speed needed to deposit hydrogel filaments of a given diameter. Finally, the proposed methodology was tested experimentally by analyzing the morphology of triple-layered square-grid hydrogel constructs printed at various applied pressures while the printing speeds matched the corresponding extrusion speeds. Taken together, the results of this study suggest that preliminary measurements and theoretical analyses can simplify the search for the optimal bioprinting parameters.
Collapse
Affiliation(s)
- Stelian Arjoca
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Center for Modeling Biological Systems and Data Analysis, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Florina Bojin
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- OncoGen Institute, 300723 Timisoara, Romania
| | - Monica Neagu
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Center for Modeling Biological Systems and Data Analysis, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Andreea Păunescu
- Carol Davila University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania
| | - Adrian Neagu
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Center for Modeling Biological Systems and Data Analysis, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Department of Physics and Astronomy, University of Missouri, Columbia, MO 65211, USA
| | - Virgil Păunescu
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- OncoGen Institute, 300723 Timisoara, Romania
| |
Collapse
|
9
|
Xiu Z, Yang Q, Xie F, Han F, He W, Liao W. Revolutionizing digestive system tumor organoids research: Exploring the potential of tumor organoids. J Tissue Eng 2024; 15:20417314241255470. [PMID: 38808253 PMCID: PMC11131411 DOI: 10.1177/20417314241255470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Digestive system tumors are the leading cause of cancer-related deaths worldwide. Despite ongoing research, our understanding of their mechanisms and treatment remain inadequate. One promising tool for clinical applications is the use of gastrointestinal tract tumor organoids, which serve as an important in vitro model. Tumor organoids exhibit a genotype similar to the patient's tumor and effectively mimic various biological processes, including tissue renewal, stem cell, and ecological niche functions, and tissue response to drugs, mutations, or injury. As such, they are valuable for drug screening, developing novel drugs, assessing patient outcomes, and supporting immunotherapy. In addition, innovative materials and techniques can be used to optimize tumor organoid culture systems. Several applications of digestive system tumor organoids have been described and have shown promising results in related aspects. In this review, we discuss the current progress, limitations, and prospects of this model for digestive system tumors.
Collapse
Affiliation(s)
- Zhian Xiu
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fusheng Xie
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Feng Han
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Weiwei He
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Weifang Liao
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
10
|
Gonzalez-Valdivieso J, Vallejo R, Rodriguez-Rojo S, Santos M, Schneider J, Arias FJ, Girotti A. CD44-targeted nanoparticles for co-delivery of docetaxel and an Akt inhibitor against colorectal cancer. BIOMATERIALS ADVANCES 2023; 154:213595. [PMID: 37639856 DOI: 10.1016/j.bioadv.2023.213595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/24/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
New strategies to develop drug-loaded nanocarriers with improved therapeutic efficacy are needed for cancer treatment. Herein we report a novel drug-delivery nanosystem comprising encapsulation of the chemotherapeutic drug docetaxel (DTX) and recombinant fusion of a small peptide inhibitor of Akt kinase within an elastin-like recombinamer (ELR) vehicle. This combined approach is also precisely targeted to colorectal cancer cells by means of a chemically conjugated DNA aptamer specific for the CD44 tumor marker. This 53 nm dual-approach nanosystem was found to selectively affect cell viability (2.5 % survival) and proliferation of colorectal cancer cells in vitro compared to endothelial cells (50 % survival), and to trigger both apoptosis- and necrosis-mediated cell death. Our findings also show that the nanohybrid particles remain stable under physiological conditions, trigger sustained drug release and possess an adequate pharmacokinetic profile after systemic intravenous administration. In vivo assays showed that these dual-approach nanohybrids significantly reduced the number of tumor polyps along the colorectal tract in a murine colorectal cancer model. Furthermore, systemic administration of advanced nanohybrids induced tissue recovery by improving the morphology of gastrointestinal crypts and the tissue architecture. Taken together, these findings indicate that our strategy of an advanced dual-approach nanosystem allows us to achieve successful controlled release of chemotherapeutics in cancer cells and may have a promising potential for colorectal cancer treatment.
Collapse
Affiliation(s)
- Juan Gonzalez-Valdivieso
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain
| | - Reinaldo Vallejo
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; BioEcoUVa, Research Institute on Bioeconomy, High Pressure Process Group, University of Valladolid, Department of Chemical Engineering and Environmental Technology, Escuela de Ingenierías Industriales, Sede Mergelina, Valladolid, Spain
| | - Soraya Rodriguez-Rojo
- BioEcoUVa, Research Institute on Bioeconomy, High Pressure Process Group, University of Valladolid, Department of Chemical Engineering and Environmental Technology, Escuela de Ingenierías Industriales, Sede Mergelina, Valladolid, Spain
| | - Mercedes Santos
- BIOFORGE Research Group (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, LUCIA Building, Valladolid, Spain
| | - Jose Schneider
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Department of Obstetrics & Gynecology, University of Valladolid, School of Medicine, Valladolid, Spain
| | - Francisco Javier Arias
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Unidad de excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), University of Valladolid CSIC, Valladolid, Spain.
| | - Alessandra Girotti
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Unidad de excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), University of Valladolid CSIC, Valladolid, Spain.
| |
Collapse
|
11
|
Minaeva ED, Antoshin AA, Kosheleva NV, Koteneva PI, Gonchukov SA, Tsypina SI, Yusupov VI, Timashev PS, Minaev NV. Laser Bioprinting with Cell Spheroids: Accurate and Gentle. MICROMACHINES 2023; 14:1152. [PMID: 37374737 DOI: 10.3390/mi14061152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023]
Abstract
Laser printing with cell spheroids can become a promising approach in tissue engineering and regenerative medicine. However, the use of standard laser bioprinters for this purpose is not optimal as they are optimized for transferring smaller objects, such as cells and microorganisms. The use of standard laser systems and protocols for the transfer of cell spheroids leads either to their destruction or to a significant deterioration in the quality of bioprinting. The possibilities of cell spheroids printing by laser-induced forward transfer in a gentle mode, which ensures good cell survival ~80% without damage and burns, were demonstrated. The proposed method showed a high spatial resolution of laser printing of cell spheroid geometric structures at the level of 62 ± 33 µm, which is significantly less than the size of the cell spheroid itself. The experiments were performed on a laboratory laser bioprinter with a sterile zone, which was supplemented with a new optical part based on the Pi-Shaper element, which allows for forming laser spots with different non-Gaussian intensity distributions. It is shown that laser spots with an intensity distribution profile of the "Two rings" type (close to Π-shaped) and a size comparable to a spheroid are optimal. To select the operating parameters of laser exposure, spheroid phantoms made of a photocurable resin and spheroids made from human umbilical cord mesenchymal stromal cells were used.
Collapse
Affiliation(s)
- Ekaterina D Minaeva
- Institute of Photon Technologies of FSRC «Crystallography and Photonics» RAS, Troitsk, 108840 Moscow, Russia
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409 Moscow, Russia
| | - Artem A Antoshin
- Institute of Photon Technologies of FSRC «Crystallography and Photonics» RAS, Troitsk, 108840 Moscow, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| | - Nastasia V Kosheleva
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
- FSBSI Institute of General Pathology and Pathophysiology, 8 Baltiyskaya, 125315 Moscow, Russia
| | - Polina I Koteneva
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| | - Sergey A Gonchukov
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409 Moscow, Russia
| | - Svetlana I Tsypina
- Institute of Photon Technologies of FSRC «Crystallography and Photonics» RAS, Troitsk, 108840 Moscow, Russia
| | - Vladimir I Yusupov
- Institute of Photon Technologies of FSRC «Crystallography and Photonics» RAS, Troitsk, 108840 Moscow, Russia
| | - Peter S Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| | - Nikita V Minaev
- Institute of Photon Technologies of FSRC «Crystallography and Photonics» RAS, Troitsk, 108840 Moscow, Russia
| |
Collapse
|
12
|
Mechanical Properties of Protein-Based Hydrogels Derived from Binary Protein Mixtures-A Feasibility Study. Polymers (Basel) 2023; 15:polym15040964. [PMID: 36850249 PMCID: PMC9964579 DOI: 10.3390/polym15040964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/08/2023] [Accepted: 02/12/2023] [Indexed: 02/18/2023] Open
Abstract
Hydrogels based on natural polymers such as proteins are considered biocompatible and, therefore, represent an interesting class of materials for application in the field of biomedicine and high-performance materials. However, there is a lack of understanding of the proteins which are able to form hydrogel networks by photoinduced dityrosine crosslinking as well as a profound knowledge of the formed network itself and the mechanisms which are responsible for the resulting mechanical properties of such protein-based hydrogels. In this study, casein, bovine serum albumin, α-amylase, and a hydrophobic elastin-like protein were used to prepare binary protein mixtures with defined concentration ratios. After polymerization, the mechanical properties of the resulting homopolymeric and copolymeric hydrogels were determined using rheological methods depending on the protein shares used. In additional uniaxial compression tests, the fracture strain was shown to be independent of the protein shares, while hydrogel toughness and compressive strength were increased for protein-based hydrogels containing casein.
Collapse
|
13
|
Abstract
Spheroids enable the study of tumors and tumor hypoxia using a more representative model of the physiological environment compared to 2D cell culture. Spheroids can be grown in a cell suspension or when adhered to a solid scaffold. The spheroid formation method used is dependent on cell type. Here we describe the most common spheroid formation methods, including hanging drop, low adhesion plates, hydrogel, micropatterned plates, and microfluidics. After spheroids are formed, they can be used for drug treatment trials and analyzed using Western Blots, qPCR, and microscopy. Microscopy can then be used to measure the invasiveness of cells when a basement membrane is added to spheroids and for monitoring changes in the proliferation, quiescent, and necrotic zones of spheroids.
Collapse
Affiliation(s)
- Sarah M Kirsh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Sydney A Pascetta
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - James Uniacke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
14
|
Hrynevich A, Li Y, Cedillo-Servin G, Malda J, Castilho M. (Bio)fabrication of microfluidic devices and organs-on-a-chip. 3D Print Med 2023. [DOI: 10.1016/b978-0-323-89831-7.00001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
15
|
Chliara MA, Elezoglou S, Zergioti I. Bioprinting on Organ-on-Chip: Development and Applications. BIOSENSORS 2022; 12:1135. [PMID: 36551101 PMCID: PMC9775862 DOI: 10.3390/bios12121135] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Organs-on-chips (OoCs) are microfluidic devices that contain bioengineered tissues or parts of natural tissues or organs and can mimic the crucial structures and functions of living organisms. They are designed to control and maintain the cell- and tissue-specific microenvironment while also providing detailed feedback about the activities that are taking place. Bioprinting is an emerging technology for constructing artificial tissues or organ constructs by combining state-of-the-art 3D printing methods with biomaterials. The utilization of 3D bioprinting and cells patterning in OoC technologies reinforces the creation of more complex structures that can imitate the functions of a living organism in a more precise way. Here, we summarize the current 3D bioprinting techniques and we focus on the advantages of 3D bioprinting compared to traditional cell seeding in addition to the methods, materials, and applications of 3D bioprinting in the development of OoC microsystems.
Collapse
Affiliation(s)
- Maria Anna Chliara
- School of Applied Mathematics and Physical Sciences, National Technical University of Athens, 15780 Zografou, Greece
- Institute of Communication and Computer Systems, 15780 Zografou, Greece
| | - Stavroula Elezoglou
- School of Applied Mathematics and Physical Sciences, National Technical University of Athens, 15780 Zografou, Greece
- PhosPrint P.C., Lefkippos Technology Park, NCSR Demokritos Patriarchou Grigoriou 5’ & Neapoleos 27, 15341 Athens, Greece
| | - Ioanna Zergioti
- School of Applied Mathematics and Physical Sciences, National Technical University of Athens, 15780 Zografou, Greece
- Institute of Communication and Computer Systems, 15780 Zografou, Greece
- PhosPrint P.C., Lefkippos Technology Park, NCSR Demokritos Patriarchou Grigoriou 5’ & Neapoleos 27, 15341 Athens, Greece
| |
Collapse
|
16
|
Damiati LA, El-Yaagoubi M, Damiati SA, Kodzius R, Sefat F, Damiati S. Role of Polymers in Microfluidic Devices. Polymers (Basel) 2022; 14:5132. [PMID: 36501526 PMCID: PMC9738615 DOI: 10.3390/polym14235132] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Polymers are sustainable and renewable materials that are in high demand due to their excellent properties. Natural and synthetic polymers with high flexibility, good biocompatibility, good degradation rate, and stiffness are widely used for various applications, such as tissue engineering, drug delivery, and microfluidic chip fabrication. Indeed, recent advances in microfluidic technology allow the fabrication of polymeric matrix to construct microfluidic scaffolds for tissue engineering and to set up a well-controlled microenvironment for manipulating fluids and particles. In this review, polymers as materials for the fabrication of microfluidic chips have been highlighted. Successful models exploiting polymers in microfluidic devices to generate uniform particles as drug vehicles or artificial cells have been also discussed. Additionally, using polymers as bioink for 3D printing or as a matrix to functionalize the sensing surface in microfluidic devices has also been mentioned. The rapid progress made in the combination of polymers and microfluidics presents a low-cost, reproducible, and scalable approach for a promising future in the manufacturing of biomimetic scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Laila A. Damiati
- Department of Biology, Collage of Science, University of Jeddah, Jeddah 23890, Saudi Arabia
| | - Marwa El-Yaagoubi
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Safa A. Damiati
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rimantas Kodzius
- Faculty of Medicine, Ludwig Maximilian University of Munich (LMU), 80539 Munich, Germany
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Farshid Sefat
- Interdisciplinary Research Centre in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford BD7 1DP, UK
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Samar Damiati
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
17
|
Johnson PA, Menegatti S, Chambers AC, Alibhai D, Collard TJ, Williams AC, Bayley H, Perriman AW. A rapid high throughput bioprinted colorectal cancer spheroid platform for in vitrodrug- and radiation-response. Biofabrication 2022; 15:014103. [PMID: 36321254 DOI: 10.1088/1758-5090/ac999f] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022]
Abstract
We describe the development of a high-throughput bioprinted colorectal cancer (CRC) spheroid platform with high levels of automation, information content, and low cell number requirement. This is achieved via the formulation of a hydrogel bioink with a compressive Young's modulus that is commensurate with that of colonic tissue (1-3 kPa), which supports exponential growth of spheroids from a wide range of CRC cell lines. The resulting spheroids display tight cell-cell junctions, bioink matrix-cell interactions and necrotic hypoxic cores. By combining high content light microscopy imaging and processing with rapid multiwell plate bioprinting, dose-response profiles are generated from CRC spheroids challenged with oxaliplatin (OX) and fluorouracil (5FU), as well as radiotherapy. Bioprinted CRC spheroids are shown to exhibit high levels of chemoresistance relative to cell monolayers, and OX was found to be significantly less effective against tumour spheroids than in monolayer culture, when compared to 5FU.
Collapse
Affiliation(s)
- Peter A Johnson
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, United Kingdom
| | - Sara Menegatti
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Adam C Chambers
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Dominic Alibhai
- Wolfson Bioimaging Facility, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Tracey J Collard
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Ann C Williams
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Hagan Bayley
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, United Kingdom
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| |
Collapse
|
18
|
Vuille-Dit-Bille E, Deshmukh DV, Connolly S, Heub S, Boder-Pasche S, Dual J, Tibbitt MW, Weder G. Tools for manipulation and positioning of microtissues. LAB ON A CHIP 2022; 22:4043-4066. [PMID: 36196619 DOI: 10.1039/d2lc00559j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Complex three-dimensional (3D) in vitro models are emerging as a key technology to support research areas in personalised medicine, such as drug development and regenerative medicine. Tools for manipulation and positioning of microtissues play a crucial role in the microtissue life cycle from production to end-point analysis. The ability to precisely locate microtissues can improve the efficiency and reliability of processes and investigations by reducing experimental time and by providing more controlled parameters. To achieve this goal, standardisation of the techniques is of primary importance. Compared to microtissue production, the field of microtissue manipulation and positioning is still in its infancy but is gaining increasing attention in the last few years. Techniques to position microtissues have been classified into four main categories: hydrodynamic techniques, bioprinting, substrate modification, and non-contact active forces. In this paper, we provide a comprehensive review of the different tools for the manipulation and positioning of microtissues that have been reported to date. The working mechanism of each technique is described, and its merits and limitations are discussed. We conclude by evaluating the potential of the different approaches to support progress in personalised medicine.
Collapse
Affiliation(s)
- Emilie Vuille-Dit-Bille
- Centre Suisse d'Electronique et de Microtechnique SA, Neuchâtel, Switzerland.
- MicroBioRobotic Systems Laboratory, Institute of Mechanical Engineering, EPFL, Lausanne, Switzerland
| | - Dhananjay V Deshmukh
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Sinéad Connolly
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zurich, Switzerland
| | - Sarah Heub
- Centre Suisse d'Electronique et de Microtechnique SA, Neuchâtel, Switzerland.
| | | | - Jürg Dual
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Gilles Weder
- Centre Suisse d'Electronique et de Microtechnique SA, Neuchâtel, Switzerland.
| |
Collapse
|
19
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|
20
|
Haas S, Desombre M, Kirschhöfer F, Huber MC, Schiller SM, Hubbuch J. Purification of a Hydrophobic Elastin-Like Protein Toward Scale-Suitable Production of Biomaterials. Front Bioeng Biotechnol 2022; 10:878838. [PMID: 35814018 PMCID: PMC9257828 DOI: 10.3389/fbioe.2022.878838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Elastin-like proteins (ELPs) are polypeptides with potential applications as renewable bio-based high-performance polymers, which undergo a stimulus-responsive reversible phase transition. The ELP investigated in this manuscript—ELP[V2Y-45]—promises fascinating mechanical properties in biomaterial applications. Purification process scalability and purification performance are important factors for the evaluation of potential industrial-scale production of ELPs. Salt-induced precipitation, inverse transition cycling (ITC), and immobilized metal ion affinity chromatography (IMAC) were assessed as purification protocols for a polyhistidine-tagged hydrophobic ELP showing low-temperature transition behavior. IMAC achieved a purity of 86% and the lowest nucleic acid contamination of all processes. Metal ion leakage did not propagate chemical modifications and could be successfully removed through size-exclusion chromatography. The simplest approach using a high-salt precipitation resulted in a 60% higher target molecule yield compared to both other approaches, with the drawback of a lower purity of 60% and higher nucleic acid contamination. An additional ITC purification led to the highest purity of 88% and high nucleic acid removal. However, expensive temperature-dependent centrifugation steps are required and aggregation effects even at low temperatures have to be considered for the investigated ELP. Therefore, ITC and IMAC are promising downstream processes for biomedical applications with scale-dependent economical costs to be considered, while salt-induced precipitation may be a fast and simple alternative for large-scale bio-based polymer production.
Collapse
Affiliation(s)
- Sandra Haas
- Institute of Process Engineering in Life Sciences, Section IV: Molecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Monika Desombre
- Institute of Process Engineering in Life Sciences, Section IV: Molecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Frank Kirschhöfer
- Institute of Functional Interfaces, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Matthias C. Huber
- Center for Biosystems Analysis, Albert‐Ludwigs‐University Freiburg, Freiburg, Germany
- Cluster of Excellence livMatS @ FIT, Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Stefan M. Schiller
- Center for Biosystems Analysis, Albert‐Ludwigs‐University Freiburg, Freiburg, Germany
- Cluster of Excellence livMatS @ FIT, Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Jürgen Hubbuch
- Institute of Process Engineering in Life Sciences, Section IV: Molecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
- Institute of Functional Interfaces, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
- *Correspondence: Jürgen Hubbuch,
| |
Collapse
|
21
|
Three-Dimensional (3D) Printing in Cancer Therapy and Diagnostics: Current Status and Future Perspectives. Pharmaceuticals (Basel) 2022; 15:ph15060678. [PMID: 35745597 PMCID: PMC9229198 DOI: 10.3390/ph15060678] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 12/10/2022] Open
Abstract
Three-dimensional (3D) printing is a technique where the products are printed layer-by-layer via a series of cross-sectional slices with the exact deposition of different cell types and biomaterials based on computer-aided design software. Three-dimensional printing can be divided into several approaches, such as extrusion-based printing, laser-induced forward transfer-based printing systems, and so on. Bio-ink is a crucial tool necessary for the fabrication of the 3D construct of living tissue in order to mimic the native tissue/cells using 3D printing technology. The formation of 3D software helps in the development of novel drug delivery systems with drug screening potential, as well as 3D constructs of tumor models. Additionally, several complex structures of inner tissues like stroma and channels of different sizes are printed through 3D printing techniques. Three-dimensional printing technology could also be used to develop therapy training simulators for educational purposes so that learners can practice complex surgical procedures. The fabrication of implantable medical devices using 3D printing technology with less risk of infections is receiving increased attention recently. A Cancer-on-a-chip is a microfluidic device that recreates tumor physiology and allows for a continuous supply of nutrients or therapeutic compounds. In this review, based on the recent literature, we have discussed various printing methods for 3D printing and types of bio-inks, and provided information on how 3D printing plays a crucial role in cancer management.
Collapse
|
22
|
Ro J, Kim J, Cho YK. Recent advances in spheroid-based microfluidic models to mimic the tumour microenvironment. Analyst 2022; 147:2023-2034. [PMID: 35485712 DOI: 10.1039/d2an00172a] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three-dimensional (3D) multicellular spheroid models can recapitulate the human tumour microenvironment with more accuracy than conventional cell culture models, as they include complex architectural structures and dynamic cellular interactions. Among the diverse platforms for spheroid formation, microfluidic platforms have been extensively applied to study spheroids because they can mimic the in vivo microenvironment. This review provides an overview of the advantages of 3D spheroid cultures with a summary of the recent applications for tumour microenvironment-focused cellular interactions, as well as the studies on spheroids and external stimuli. These 3D tumour spheroid-based microfluidic devices will provide a platform for a better understanding of cellular and external interactions, as well as the discovery of cancer therapeutics.
Collapse
Affiliation(s)
- Jooyoung Ro
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Korea. .,Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Korea
| | - Junyoung Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Korea.
| | - Yoon-Kyoung Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Korea. .,Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Korea
| |
Collapse
|
23
|
Affiliation(s)
| | - Brian R. James
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
24
|
|
25
|
Schumacher L, Siemsen K, Appiah C, Rajput S, Heitmann A, Selhuber-Unkel C, Staubitz A. A Co-Polymerizable Linker for the Covalent Attachment of Fibronectin Makes pHEMA Hydrogels Cell-Adhesive. Gels 2022; 8:258. [PMID: 35621556 PMCID: PMC9140594 DOI: 10.3390/gels8050258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 11/20/2022] Open
Abstract
Hydrogels are attractive biomaterials because their chemical and mechanical properties can be tailored to mimic those of biological tissues. However, many hydrogels do not allow cell or protein attachment. Therefore, they are post-synthetically functionalized by adding functional groups for protein binding, which then allows cell adhesion in cell culture substrates. However, the degree of functionalization and covalent binding is difficult to analyze in these cases. Moreover, the density of the functional groups and the homogeneity of their distribution is hard to control. This work introduces another strategy for the biofunctionalization of hydrogels: we synthesized a polymerizable linker that serves as a direct junction between the polymeric structure and cell adhesion proteins. This maleimide-containing, polymerizable bio-linker was copolymerized with non-functionalized monomers to produce a bioactive hydrogel based on poly(2-hydroxyethyl methacrylate) (pHEMA). Therefore, the attachment site was only controlled by the polymerization process and was thus uniformly distributed throughout the hydrogel. In this way, the bio-conjugation by a protein-binding thiol-maleimide Michael-type reaction was possible in the entire hydrogel matrix. This approach enabled a straightforward and highly effective biofunctionalization of pHEMA with the adhesion protein fibronectin. The bioactivity of the materials was demonstrated by the successful adhesion of fibroblast cells.
Collapse
Affiliation(s)
- Laura Schumacher
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Straße 7, D-28359 Bremen, Germany; (L.S.); (C.A.); (A.H.)
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstr. 1, D-28359 Bremen, Germany
| | - Katharina Siemsen
- Biocompatible Nanomaterials, Institute for Materials Science, Kiel University, Kaiserstr. 2, D-24143 Kiel, Germany;
| | - Clement Appiah
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Straße 7, D-28359 Bremen, Germany; (L.S.); (C.A.); (A.H.)
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstr. 1, D-28359 Bremen, Germany
| | - Sunil Rajput
- Institute for Molecular Systems Engineering (IMSE), Heidelberg University, INF 253, D-69120 Heidelberg, Germany;
| | - Anne Heitmann
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Straße 7, D-28359 Bremen, Germany; (L.S.); (C.A.); (A.H.)
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstr. 1, D-28359 Bremen, Germany
| | - Christine Selhuber-Unkel
- Institute for Molecular Systems Engineering (IMSE), Heidelberg University, INF 253, D-69120 Heidelberg, Germany;
- Max Planck School Matter to Life, Jahnstraße 29, D-69120 Heidelberg, Germany
| | - Anne Staubitz
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Straße 7, D-28359 Bremen, Germany; (L.S.); (C.A.); (A.H.)
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstr. 1, D-28359 Bremen, Germany
| |
Collapse
|
26
|
Ze Y, Li Y, Huang L, Shi Y, Li P, Gong P, Lin J, Yao Y. Biodegradable Inks in Indirect Three-Dimensional Bioprinting for Tissue Vascularization. Front Bioeng Biotechnol 2022; 10:856398. [PMID: 35402417 PMCID: PMC8990266 DOI: 10.3389/fbioe.2022.856398] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/09/2022] [Indexed: 02/05/2023] Open
Abstract
Mature vasculature is important for the survival of bioengineered tissue constructs, both in vivo and in vitro; however, the fabrication of fully vascularized tissue constructs remains a great challenge in tissue engineering. Indirect three-dimensional (3D) bioprinting refers to a 3D printing technique that can rapidly fabricate scaffolds with controllable internal pores, cavities, and channels through the use of sacrificial molds. It has attracted much attention in recent years owing to its ability to create complex vascular network-like channels through thick tissue constructs while maintaining endothelial cell activity. Biodegradable materials play a crucial role in tissue engineering. Scaffolds made of biodegradable materials act as temporary templates, interact with cells, integrate with native tissues, and affect the results of tissue remodeling. Biodegradable ink selection, especially the choice of scaffold and sacrificial materials in indirect 3D bioprinting, has been the focus of several recent studies. The major objective of this review is to summarize the basic characteristics of biodegradable materials commonly used in indirect 3D bioprinting for vascularization, and to address recent advances in applying this technique to the vascularization of different tissues. Furthermore, the review describes how indirect 3D bioprinting creates blood vessels and vascularized tissue constructs by introducing the methodology and biodegradable ink selection. With the continuous improvement of biodegradable materials in the future, indirect 3D bioprinting will make further contributions to the development of this field.
Collapse
Affiliation(s)
- Yiting Ze
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanxi Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linyang Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yixin Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiran Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Salar Amoli M, EzEldeen M, Jacobs R, Bloemen V. Materials for Dentoalveolar Bioprinting: Current State of the Art. Biomedicines 2021; 10:biomedicines10010071. [PMID: 35052751 PMCID: PMC8773444 DOI: 10.3390/biomedicines10010071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/25/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Although current treatments can successfully address a wide range of complications in the dentoalveolar region, they often still suffer from drawbacks and limitations, resulting in sub-optimal treatments for specific problems. In recent decades, significant progress has been made in the field of tissue engineering, aiming at restoring damaged tissues via a regenerative approach. Yet, the translation into a clinical product is still challenging. Novel technologies such as bioprinting have been developed to solve some of the shortcomings faced in traditional tissue engineering approaches. Using automated bioprinting techniques allows for precise placement of cells and biological molecules and for geometrical patient-specific design of produced biological scaffolds. Recently, bioprinting has also been introduced into the field of dentoalveolar tissue engineering. However, the choice of a suitable material to encapsulate cells in the development of so-called bioinks for bioprinting dentoalveolar tissues is still a challenge, considering the heterogeneity of these tissues and the range of properties they possess. This review, therefore, aims to provide an overview of the current state of the art by discussing the progress of the research on materials used for dentoalveolar bioprinting, highlighting the advantages and shortcomings of current approaches and considering opportunities for further research.
Collapse
Affiliation(s)
- Mehdi Salar Amoli
- Surface and Interface Engineered Materials (SIEM), Campus Group T, KU Leuven, Andreas Vesaliusstraat 13, 3000 Leuven, Belgium;
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven and Oral and Maxillofacial Surgery, University Hospitals Leuven, Kapucijnenvoer 33, 3000 Leuven, Belgium; (M.E.); (R.J.)
| | - Mostafa EzEldeen
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven and Oral and Maxillofacial Surgery, University Hospitals Leuven, Kapucijnenvoer 33, 3000 Leuven, Belgium; (M.E.); (R.J.)
- Department of Oral Health Sciences, KU Leuven and Paediatric Dentistry and Special Dental Care, University Hospitals Leuven, Kapucijnenvoer 33, 3000 Leuven, Belgium
| | - Reinhilde Jacobs
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven and Oral and Maxillofacial Surgery, University Hospitals Leuven, Kapucijnenvoer 33, 3000 Leuven, Belgium; (M.E.); (R.J.)
- Department of Dental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Veerle Bloemen
- Surface and Interface Engineered Materials (SIEM), Campus Group T, KU Leuven, Andreas Vesaliusstraat 13, 3000 Leuven, Belgium;
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Correspondence: ; Tel.: +32-16-30-10-95
| |
Collapse
|
28
|
Dai M, Belaïdi JP, Fleury G, Garanger E, Rielland M, Schultze X, Lecommandoux S. Elastin-like Polypeptide-Based Bioink: A Promising Alternative for 3D Bioprinting. Biomacromolecules 2021; 22:4956-4966. [PMID: 34751573 DOI: 10.1021/acs.biomac.1c00861] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Three-dimensional (3D) bioprinting offers a great alternative to traditional techniques in tissue reconstruction, based on seeding cells manually into a scaffold, to better reproduce organs' complexity. When a suitable bioink is engineered with appropriate physicochemical properties, such a process can advantageously provide a spatial control of the patterning that improves tissue reconstruction. The design of an adequate bioink must fulfill a long list of criteria including biocompatibility, printability, and stability. In this context, we have developed a bioink containing a precisely controlled recombinant biopolymer, namely, elastin-like polypeptide (ELP). This material was further chemoselectively modified with cross-linkable moieties to provide a 3D network through photopolymerization. ELP chains were additionally either functionalized with a peptide sequence Gly-Arg-Gly-Asp-Ser (GRGDS) or combined with collagen I to enable cell adhesion. Our ELP-based bioinks were found to be printable, while providing excellent mechanical properties such as stiffness and elasticity in their cross-linked form. Besides, they were demonstrated to be biocompatible, showing viability and adhesion of dermal normal human fibroblasts (NHF). Expressions of specific extracellular matrix (ECM) protein markers as pro-collagen I, elastin, fibrillin, and fibronectin were revealed within the 3D network containing cells after only 18 days of culture, showing the great potential of ELP-based bioinks for tissue engineering.
Collapse
Affiliation(s)
- Michèle Dai
- L'Oréal Recherche Avancée, 1 avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France.,Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | - Jean-Philippe Belaïdi
- L'Oréal Recherche Avancée, 1 avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | - Guillaume Fleury
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | - Elisabeth Garanger
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | - Maïté Rielland
- L'Oréal Recherche Avancée, 1 avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | - Xavier Schultze
- L'Oréal Recherche Avancée, 1 avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | | |
Collapse
|
29
|
Zhuang P, Chiang YH, Fernanda MS, He M. Using Spheroids as Building Blocks Towards 3D Bioprinting of Tumor Microenvironment. Int J Bioprint 2021; 7:444. [PMID: 34805601 PMCID: PMC8600307 DOI: 10.18063/ijb.v7i4.444] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer still ranks as a leading cause of mortality worldwide. Although considerable efforts have been dedicated to anticancer therapeutics, progress is still slow, partially due to the absence of robust prediction models. Multicellular tumor spheroids, as a major three-dimensional (3D) culture model exhibiting features of avascular tumors, gained great popularity in pathophysiological studies and high throughput drug screening. However, limited control over cellular and structural organization is still the key challenge in achieving in vivo like tissue microenvironment. 3D bioprinting has made great strides toward tissue/organ mimicry, due to its outstanding spatial control through combining both cells and materials, scalability, and reproducibility. Prospectively, harnessing the power from both 3D bioprinting and multicellular spheroids would likely generate more faithful tumor models and advance our understanding on the mechanism of tumor progression. In this review, the emerging concept on using spheroids as a building block in 3D bioprinting for tumor modeling is illustrated. We begin by describing the context of the tumor microenvironment, followed by an introduction of various methodologies for tumor spheroid formation, with their specific merits and drawbacks. Thereafter, we present an overview of existing 3D printed tumor models using spheroids as a focus. We provide a compilation of the contemporary literature sources and summarize the overall advancements in technology and possibilities of using spheroids as building blocks in 3D printed tissue modeling, with a particular emphasis on tumor models. Future outlooks about the wonderous advancements of integrated 3D spheroidal printing conclude this review.
Collapse
Affiliation(s)
- Pei Zhuang
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, 32610, USA
| | - Yi-Hua Chiang
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, 32610, USA
| | | | - Mei He
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, 32610, USA
| |
Collapse
|
30
|
Parisi C, Qin K, Fernandes FM. Colonization versus encapsulation in cell-laden materials design: porosity and process biocompatibility determine cellularization pathways. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2021; 379:20200344. [PMID: 34334019 DOI: 10.1098/rsta.2020.0344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/28/2021] [Indexed: 06/13/2023]
Abstract
Seeding materials with living cells has been-and still is-one of the most promising approaches to reproduce the complexity and the functionality of living matter. The strategies to associate living cells with materials are limited to cell encapsulation and colonization, however, the requirements for these two approaches have been seldom discussed systematically. Here we propose a simple two-dimensional map based on materials' pore size and the cytocompatibility of their fabrication process to draw, for the first time, a guide to building cellularized materials. We believe this approach may serve as a straightforward guideline to design new, more relevant materials, able to seize the complexity and the function of biological materials. This article is part of the theme issue 'Bio-derived and bioinspired sustainable advanced materials for emerging technologies (part 1)'.
Collapse
Affiliation(s)
- Cleo Parisi
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| | - Kankan Qin
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| | - Francisco M Fernandes
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| |
Collapse
|
31
|
Kim J, Jang J, Cho DW. Recapitulating the Cancer Microenvironment Using Bioprinting Technology for Precision Medicine. MICROMACHINES 2021; 12:1122. [PMID: 34577765 PMCID: PMC8472267 DOI: 10.3390/mi12091122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 12/20/2022]
Abstract
The complex and heterogenous nature of cancer contributes to the development of cancer cell drug resistance. The construction of the cancer microenvironment, including the cell-cell interactions and extracellular matrix (ECM), plays a significant role in the development of drug resistance. Traditional animal models used in drug discovery studies have been associated with feasibility issues that limit the recapitulation of human functions; thus, in vitro models have been developed to reconstruct the human cancer system. However, conventional two-dimensional and three-dimensional (3D) in vitro cancer models are limited in their ability to emulate complex cancer microenvironments. Advances in technologies, including bioprinting and cancer microenvironment reconstruction, have demonstrated the potential to overcome some of the limitations of conventional models. This study reviews some representative bioprinted in vitro models used in cancer research, particularly fabrication strategies for modeling and consideration of essential factors needed for the reconstruction of the cancer microenvironment. In addition, we highlight recent studies that applied such models, including application in precision medicine using advanced bioprinting technologies to fabricate biomimetic cancer models. Furthermore, we discuss current challenges in 3D bioprinting and suggest possible strategies to construct in vitro models that better mimic the pathophysiology of the cancer microenvironment for application in clinical settings.
Collapse
Affiliation(s)
- Jisoo Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
| | - Dong-Woo Cho
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
32
|
Veiga A, Silva IV, Duarte MM, Oliveira AL. Current Trends on Protein Driven Bioinks for 3D Printing. Pharmaceutics 2021; 13:1444. [PMID: 34575521 PMCID: PMC8471984 DOI: 10.3390/pharmaceutics13091444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023] Open
Abstract
In the last decade, three-dimensional (3D) extrusion bioprinting has been on the top trend for innovative technologies in the field of biomedical engineering. In particular, protein-based bioinks such as collagen, gelatin, silk fibroin, elastic, fibrin and protein complexes based on decellularized extracellular matrix (dECM) are receiving increasing attention. This current interest is the result of protein's tunable properties, biocompatibility, environmentally friendly nature and possibility to provide cells with the adequate cues, mimicking the extracellular matrix's function. In this review we describe the most relevant stages of the development of a protein-driven bioink. The most popular formulations, molecular weights and extraction methods are covered. The different crosslinking methods used in protein bioinks, the formulation with other polymeric systems or molecules of interest as well as the bioprinting settings are herein highlighted. The cell embedding procedures, the in vitro, in vivo, in situ studies and final applications are also discussed. Finally, we approach the development and optimization of bioinks from a sequential perspective, discussing the relevance of each parameter during the pre-processing, processing, and post-processing stages of technological development. Through this approach the present review expects to provide, in a sequential manner, helpful methodological guidelines for the development of novel bioinks.
Collapse
Affiliation(s)
- Anabela Veiga
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, 4099-002 Porto, Portugal
| | - Inês V. Silva
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
| | - Marta M. Duarte
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
| | - Ana L. Oliveira
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
| |
Collapse
|
33
|
Wang B, Patkar SS, Kiick KL. Application of Thermoresponsive Intrinsically Disordered Protein Polymers in Nanostructured and Microstructured Materials. Macromol Biosci 2021; 21:e2100129. [PMID: 34145967 PMCID: PMC8449816 DOI: 10.1002/mabi.202100129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/25/2021] [Indexed: 01/15/2023]
Abstract
Modulation of inter- and intramolecular interactions between bioinspired designer molecules can be harnessed for developing functional structures that mimic the complex hierarchical organization of multicomponent assemblies observed in nature. Furthermore, such multistimuli-responsive molecules offer orthogonal tunability for generating versatile multifunctional platforms via independent biochemical and biophysical cues. In this review, the remarkable physicochemical and mechanical properties of genetically engineered protein polymers derived from intrinsically disordered proteins, specifically elastin and resilin, are discussed. This review highlights emerging technologies which use them as building blocks in the fabrication of highly programmable structured biomaterials for applications in delivery of biotherapeutic cargo and regenerative medicine.
Collapse
Affiliation(s)
- Bin Wang
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
| | - Sai S Patkar
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Laboratory, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, Ammon Pinizzotto Biopharmaceutical Innovation Center, 590 Avenue 1743, Newark, DE, 19713, USA
| |
Collapse
|
34
|
Jeon S, Lee SH, Ahmed SB, Han J, Heo SJ, Kang HW. 3D cell aggregate printing technology and its applications. Essays Biochem 2021; 65:467-480. [PMID: 34223609 PMCID: PMC11293493 DOI: 10.1042/ebc20200128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/14/2021] [Accepted: 06/08/2021] [Indexed: 12/27/2022]
Abstract
Various cell aggregate culture technologies have been developed and actively applied to tissue engineering and organ-on-a-chip. However, the conventional culture technologies are labor-intensive, and their outcomes are highly user dependent. In addition, the technologies cannot be used to produce three-dimensional (3D) complex tissues. In this regard, 3D cell aggregate printing technology has attracted increased attention from many researchers owing to its 3D processability. The technology allows the fabrication of 3D freeform constructs using multiple types of cell aggregates in an automated manner. Technological advancement has resulted in the development of a printing technology with a high resolution of approximately 20 μm in 3D space. A high-speed printing technology that can print a cell aggregate in milliseconds has also been introduced. The developed aggregate printing technologies are being actively applied to produce various types of engineered tissues. Although various types of high-performance printing technologies have been developed, there are still some technical obstacles in the fabrication of engineered tissues that mimic the structure and function of native tissues. This review highlights the central importance and current technical level of 3D cell aggregate printing technology, and their applications to tissue/disease models, artificial tissues, and drug-screening platforms. The paper also discusses the remaining hurdles and future directions of the printing processes.
Collapse
Affiliation(s)
- Seunggyu Jeon
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
| | - Se-Hwan Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
- Department of Orthopaedic Surgery, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, U.S.A
| | - Saeed B. Ahmed
- Department of Orthopaedic Surgery, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, U.S.A
| | - Jonghyeuk Han
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
| | - Su-Jin Heo
- Department of Orthopaedic Surgery, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, U.S.A
| | - Hyun-Wook Kang
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
| |
Collapse
|
35
|
Mansouri M, Leipzig ND. Advances in removing mass transport limitations for more physiologically relevant in vitro 3D cell constructs. BIOPHYSICS REVIEWS 2021; 2:021305. [PMID: 38505119 PMCID: PMC10903443 DOI: 10.1063/5.0048837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/31/2021] [Indexed: 03/21/2024]
Abstract
Spheroids and organoids are promising models for biomedical applications ranging from human disease modeling to drug discovery. A main goal of these 3D cell-based platforms is to recapitulate important physiological parameters of their in vivo organ counterparts. One way to achieve improved biomimetic architectures and functions is to culture cells at higher density and larger total numbers. However, poor nutrient and waste transport lead to low stability, survival, and functionality over extended periods of time, presenting outstanding challenges in this field. Fortunately, important improvements in culture strategies have enhanced the survival and function of cells within engineered microtissues/organs. Here, we first discuss the challenges of growing large spheroids/organoids with a focus on mass transport limitations, then highlight recent tools and methodologies that are available for producing and sustaining functional 3D in vitro models. This information points toward the fact that there is a critical need for the continued development of novel cell culture strategies that address mass transport in a physiologically relevant human setting to generate long-lasting and large-sized spheroids/organoids.
Collapse
Affiliation(s)
- Mona Mansouri
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| | - Nic D. Leipzig
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| |
Collapse
|
36
|
Chen EP, Toksoy Z, Davis BA, Geibel JP. 3D Bioprinting of Vascularized Tissues for in vitro and in vivo Applications. Front Bioeng Biotechnol 2021; 9:664188. [PMID: 34055761 PMCID: PMC8158943 DOI: 10.3389/fbioe.2021.664188] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 12/23/2022] Open
Abstract
With a limited supply of organ donors and available organs for transplantation, the aim of tissue engineering with three-dimensional (3D) bioprinting technology is to construct fully functional and viable tissue and organ replacements for various clinical applications. 3D bioprinting allows for the customization of complex tissue architecture with numerous combinations of materials and printing methods to build different tissue types, and eventually fully functional replacement organs. The main challenge of maintaining 3D printed tissue viability is the inclusion of complex vascular networks for nutrient transport and waste disposal. Rapid development and discoveries in recent years have taken huge strides toward perfecting the incorporation of vascular networks in 3D printed tissue and organs. In this review, we will discuss the latest advancements in fabricating vascularized tissue and organs including novel strategies and materials, and their applications. Our discussion will begin with the exploration of printing vasculature, progress through the current statuses of bioprinting tissue/organoids from bone to muscles to organs, and conclude with relevant applications for in vitro models and drug testing. We will also explore and discuss the current limitations of vascularized tissue engineering and some of the promising future directions this technology may bring.
Collapse
Affiliation(s)
- Earnest P Chen
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Yale College, Yale University, New Haven, CT, United States
| | - Zeren Toksoy
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Yale College, Yale University, New Haven, CT, United States
| | - Bruce A Davis
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT, United States
| | - John P Geibel
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
37
|
Mattei F, Andreone S, Mencattini A, De Ninno A, Businaro L, Martinelli E, Schiavoni G. Oncoimmunology Meets Organs-on-Chip. Front Mol Biosci 2021; 8:627454. [PMID: 33842539 PMCID: PMC8032996 DOI: 10.3389/fmolb.2021.627454] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/04/2021] [Indexed: 01/04/2023] Open
Abstract
Oncoimmunology represents a biomedical research discipline coined to study the roles of immune system in cancer progression with the aim of discovering novel strategies to arm it against the malignancy. Infiltration of immune cells within the tumor microenvironment is an early event that results in the establishment of a dynamic cross-talk. Here, immune cells sense antigenic cues to mount a specific anti-tumor response while cancer cells emanate inhibitory signals to dampen it. Animals models have led to giant steps in this research context, and several tools to investigate the effect of immune infiltration in the tumor microenvironment are currently available. However, the use of animals represents a challenge due to ethical issues and long duration of experiments. Organs-on-chip are innovative tools not only to study how cells derived from different organs interact with each other, but also to investigate on the crosstalk between immune cells and different types of cancer cells. In this review, we describe the state-of-the-art of microfluidics and the impact of OOC in the field of oncoimmunology underlining the importance of this system in the advancements on the complexity of tumor microenvironment.
Collapse
Affiliation(s)
- Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Arianna Mencattini
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy.,Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (ICLOC), University of Rome Tor Vergata, Rome, Italy
| | - Adele De Ninno
- Institute for Photonics and Nanotechnologies, Italian National Research Council, Rome, Italy
| | - Luca Businaro
- Institute for Photonics and Nanotechnologies, Italian National Research Council, Rome, Italy
| | - Eugenio Martinelli
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy.,Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (ICLOC), University of Rome Tor Vergata, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
38
|
Tamay DG, Hasirci N. Bioinks-materials used in printing cells in designed 3D forms. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1072-1106. [PMID: 33720806 DOI: 10.1080/09205063.2021.1892470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Use of materials to activate non-functional or damaged organs and tissues goes back to early ages. The first materials used for this purpose were metals, and in time, novel materials such as ceramics, polymers and composites were introduced to the field to serve in medical applications. In the last decade, the advances in material sciences, cell biology, technology and engineering made 3D printing of living tissues or organ models in the designed structure and geometry possible by using cells alone or together with hydrogels through additive manufacturing. This review aims to give a brief information about the chemical structures and properties of bioink materials and their applications in the production of 3D tissue constructs.
Collapse
Affiliation(s)
- Dilara Goksu Tamay
- BIOMATEN - Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, Turkey.,Department of Biomedical Engineering, Middle East Technical University, Ankara, Turkey
| | - Nesrin Hasirci
- BIOMATEN - Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, Turkey.,Department of Biomedical Engineering, Middle East Technical University, Ankara, Turkey.,Department of Chemistry, Middle East Technical University, Ankara, Turkey.,Tissue Engineering and Biomaterial Research Center, Near East University, TRNC, Mersin 10, Turkey
| |
Collapse
|
39
|
Sánchez-Salazar MG, Álvarez MM, Trujillo-de Santiago G. Advances in 3D bioprinting for the biofabrication of tumor models. ACTA ACUST UNITED AC 2021. [DOI: 10.1016/j.bprint.2020.e00120] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
40
|
Hull SM, Lindsay CD, Brunel LG, Shiwarski DJ, Tashman JW, Roth JG, Myung D, Feinberg AW, Heilshorn SC. 3D Bioprinting using UNIversal Orthogonal Network (UNION) Bioinks. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007983. [PMID: 33613150 PMCID: PMC7888563 DOI: 10.1002/adfm.202007983] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Indexed: 05/02/2023]
Abstract
Three-dimensional (3D) bioprinting is a promising technology to produce tissue-like structures, but a lack of diversity in bioinks is a major limitation. Ideally each cell type would be printed in its own customizable bioink. To fulfill this need for a universally applicable bioink strategy, we developed a versatile, bioorthogonal bioink crosslinking mechanism that is cell compatible and works with a range of polymers. We term this family of materials UNIversal, Orthogonal Network (UNION) bioinks. As demonstration of UNION bioink versatility, gelatin, hyaluronic acid (HA), recombinant elastin-like protein (ELP), and polyethylene glycol (PEG) were each used as backbone polymers to create inks with storage moduli spanning 200 to 10,000 Pa. Because UNION bioinks are crosslinked by a common chemistry, multiple materials can be printed together to form a unified, cohesive structure. This approach is compatible with any support bath that enables diffusion of UNION crosslinkers. Both matrix-adherent human corneal mesenchymal stromal cells and non-matrix-adherent human induced pluripotent stem cell-derived neural progenitor spheroids were printed with UNION bioinks. The cells retained high viability and expressed characteristic phenotypic markers after printing. Thus, UNION bioinks are a versatile strategy to expand the toolkit of customizable materials available for 3D bioprinting.
Collapse
Affiliation(s)
- Sarah M Hull
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Christopher D Lindsay
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Daniel J Shiwarski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Joshua W Tashman
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Julien G Roth
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - David Myung
- Department of Ophthalmology, Stanford University, Stanford, CA 94305, USA
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Sarah C Heilshorn
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
41
|
Choi YJ, Park H, Ha DH, Yun HS, Yi HG, Lee H. 3D Bioprinting of In Vitro Models Using Hydrogel-Based Bioinks. Polymers (Basel) 2021; 13:366. [PMID: 33498852 PMCID: PMC7865738 DOI: 10.3390/polym13030366] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), which has recently emerged as a global pandemic, has caused a serious economic crisis due to the social disconnection and physical distancing in human society. To rapidly respond to the emergence of new diseases, a reliable in vitro model needs to be established expeditiously for the identification of appropriate therapeutic agents. Such models can be of great help in validating the pathological behavior of pathogens and therapeutic agents. Recently, in vitro models representing human organs and tissues and biological functions have been developed based on high-precision 3D bioprinting. In this paper, we delineate an in-depth assessment of the recently developed 3D bioprinting technology and bioinks. In particular, we discuss the latest achievements and future aspects of the use of 3D bioprinting for in vitro modeling.
Collapse
Affiliation(s)
- Yeong-Jin Choi
- Department of Advanced Biomaterials Research, Korea Institute of Materials Science (KIMS), 797, Changwon 51508, Korea; (Y.-J.C.); (H.P.); (H.-S.Y.)
| | - Honghyun Park
- Department of Advanced Biomaterials Research, Korea Institute of Materials Science (KIMS), 797, Changwon 51508, Korea; (Y.-J.C.); (H.P.); (H.-S.Y.)
| | | | - Hui-Suk Yun
- Department of Advanced Biomaterials Research, Korea Institute of Materials Science (KIMS), 797, Changwon 51508, Korea; (Y.-J.C.); (H.P.); (H.-S.Y.)
| | - Hee-Gyeong Yi
- Department of Rural and Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University (CNU), Gwangju 61186, Korea
| | - Hyungseok Lee
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea
- Interdisciplinary Program in Biohealth-Machinery Convergence Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea
| |
Collapse
|
42
|
Tan B, Gan S, Wang X, Liu W, Li X. Applications of 3D bioprinting in tissue engineering: advantages, deficiencies, improvements, and future perspectives. J Mater Chem B 2021; 9:5385-5413. [PMID: 34124724 DOI: 10.1039/d1tb00172h] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the past decade, 3D bioprinting technology has progressed tremendously in the field of tissue engineering in its ability to fabricate individualized biological constructs with precise geometric designability, which offers us the capability to bridge the divergence between engineered tissue constructs and natural tissues. In this work, we first review the current widely used 3D bioprinting approaches, cells, and materials. Next, the updated applications of this technique in tissue engineering, including bone tissue, cartilage tissue, vascular grafts, skin, neural tissue, heart tissue, liver tissue and lung tissue, are briefly introduced. Then, the prominent advantages of 3D bioprinting in tissue engineering are summarized in detail: rapidly prototyping the customized structure, delivering cell-laden materials with high precision in space, and engineering with a highly controllable microenvironment. The current technical deficiencies of 3D bioprinted constructs in terms of mechanical properties and cell behaviors are afterward illustrated, as well as corresponding improvements. Finally, we conclude with future perspectives about 3D bioprinting in tissue engineering.
Collapse
Affiliation(s)
- Baosen Tan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Shaolei Gan
- Jiangxi Borayer Biotech Co., Ltd, Nanchang 330052, China
| | - Xiumei Wang
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Wenyong Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| |
Collapse
|
43
|
Duarte Campos DF, De Laporte L. Digitally Fabricated and Naturally Augmented In Vitro Tissues. Adv Healthc Mater 2021; 10:e2001253. [PMID: 33191651 PMCID: PMC11468916 DOI: 10.1002/adhm.202001253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/04/2020] [Indexed: 01/29/2023]
Abstract
Human in vitro tissues are extracorporeal 3D cultures of human cells embedded in biomaterials, commonly hydrogels, which recapitulate the heterogeneous, multiscale, and architectural environment of the human body. Contemporary strategies used in 3D tissue and organ engineering integrate the use of automated digital manufacturing methods, such as 3D printing, bioprinting, and biofabrication. Human tissues and organs, and their intra- and interphysiological interplay, are particularly intricate. For this reason, attentiveness is rising to intersect materials science, medicine, and biology with arts and informatics. This report presents advances in computational modeling of bioink polymerization and its compatibility with bioprinting, the use of digital design and fabrication in the development of fluidic culture devices, and the employment of generative algorithms for modeling the natural and biological augmentation of in vitro tissues. As a future direction, the use of serially linked in vitro tissues as human body-mimicking systems and their application in drug pharmacokinetics and metabolism, disease modeling, and diagnostics are discussed.
Collapse
Affiliation(s)
- Daniela F. Duarte Campos
- Department of Advanced Materials for BiomedicineInstitute of Applied Medical EngineeringRWTH Aachen UniversityAachen52074Germany
| | - Laura De Laporte
- Department of Advanced Materials for BiomedicineInstitute of Applied Medical EngineeringRWTH Aachen UniversityAachen52074Germany
- DWI—Leibniz Institute for Interactive MaterialsAachen52074Germany
- Department of Technical and Macromolecular ChemistryRWTH Aachen UniversityAachen52074Germany
| |
Collapse
|