1
|
Bardini R, Di Carlo S. Computational methods for biofabrication in tissue engineering and regenerative medicine - a literature review. Comput Struct Biotechnol J 2024; 23:601-616. [PMID: 38283852 PMCID: PMC10818159 DOI: 10.1016/j.csbj.2023.12.035] [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: 08/31/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/30/2024] Open
Abstract
This literature review rigorously examines the growing scientific interest in computational methods for Tissue Engineering and Regenerative Medicine biofabrication, a leading-edge area in biomedical innovation, emphasizing the need for accurate, multi-stage, and multi-component biofabrication process models. The paper presents a comprehensive bibliometric and contextual analysis, followed by a literature review, to shed light on the vast potential of computational methods in this domain. It reveals that most existing methods focus on single biofabrication process stages and components, and there is a significant gap in approaches that utilize accurate models encompassing both biological and technological aspects. This analysis underscores the indispensable role of these methods in understanding and effectively manipulating complex biological systems and the necessity for developing computational methods that span multiple stages and components. The review concludes that such comprehensive computational methods are essential for developing innovative and efficient Tissue Engineering and Regenerative Medicine biofabrication solutions, driving forward advancements in this dynamic and evolving field.
Collapse
Affiliation(s)
- Roberta Bardini
- Department of Control and Computer Engineering, Polytechnic University of Turin, Corso Duca Degli Abruzzi, 24, Turin, 10129, Italy
| | - Stefano Di Carlo
- Department of Control and Computer Engineering, Polytechnic University of Turin, Corso Duca Degli Abruzzi, 24, Turin, 10129, Italy
| |
Collapse
|
2
|
Govender M, Indermun S, Choonara YE. 3D bioprinted microneedles: merging drug delivery and scaffold science for tissue-specific applications. Expert Opin Drug Deliv 2024; 21:1559-1572. [PMID: 38722022 DOI: 10.1080/17425247.2024.2351928] [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: 02/15/2024] [Accepted: 05/02/2024] [Indexed: 11/10/2024]
Abstract
INTRODUCTION Three-Dimensional (3D) microneedles have recently gained significant attention due to their versatility, biocompatibility, enhanced permeation, and predictable behavior. The incorporation of biological agents into these 3D constructs has advanced the traditional microneedle into an effective platform for wide-ranging applications. AREAS COVERED This review discusses the current state of microneedle fabrication as well as the developed 3D printed microneedles incorporating labile pharmaceutical agents and biological materials for potential biomedical applications. The mechanical and processing considerations for the preparation of microneedles and the barriers to effective 3D printing of microneedle constructs have additionally been reviewed along with their therapeutic applications and potential for tissue engineering and regenerative applications. Additionally, the regulatory considerations for microneedle approval have been discussed as well as the current clinical trial and patent landscapes. EXPERT OPINION The fields of tissue engineering and regenerative medicine are evolving at a significant pace with researchers constantly focused on incorporating advanced manufacturing techniques for the development of versatile, complex, and biologically specific platforms. 3D bioprinted microneedles, fabricated using conventional 3D printing techniques, have resultantly provided an alternative to 2D bioscaffolds through the incorporation of biological materials within 3D constructs while providing further mechanical stability, increased bioactive permeation and improved innervation into surrounding tissues. This advancement therefore potentially allows for a more effective biomimetic construct with improved tissue-specific cellular growth for the enhanced treatment of physiological conditions requiring tissue regeneration and replacement.
Collapse
Affiliation(s)
- Mershen Govender
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Sunaina Indermun
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| |
Collapse
|
3
|
Garciamendez-Mijares CE, Aguilar FJ, Hernandez P, Kuang X, Gonzalez M, Ortiz V, Riesgo RA, Ruiz DSR, Rivera VAM, Rodriguez JC, Mestre FL, Castillo PC, Perez A, Cruz LM, Lim KS, Zhang YS. Design considerations for digital light processing bioprinters. APPLIED PHYSICS REVIEWS 2024; 11:031314. [PMID: 39221036 PMCID: PMC11284760 DOI: 10.1063/5.0187558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 07/02/2024] [Indexed: 09/04/2024]
Abstract
With the rapid development and popularization of additive manufacturing, different technologies, including, but not limited to, extrusion-, droplet-, and vat-photopolymerization-based fabrication techniques, have emerged that have allowed tremendous progress in three-dimensional (3D) printing in the past decades. Bioprinting, typically using living cells and/or biomaterials conformed by different printing modalities, has produced functional tissues. As a subclass of vat-photopolymerization bioprinting, digital light processing (DLP) uses digitally controlled photomasks to selectively solidify liquid photocurable bioinks to construct complex physical objects in a layer-by-layer manner. DLP bioprinting presents unique advantages, including short printing times, relatively low manufacturing costs, and decently high resolutions, allowing users to achieve significant progress in the bioprinting of tissue-like complex structures. Nevertheless, the need to accommodate different materials while bioprinting and improve the printing performance has driven the rapid progress in DLP bioprinters, which requires multiple pieces of knowledge ranging from optics, electronics, software, and materials beyond the biological aspects. This raises the need for a comprehensive review to recapitulate the most important considerations in the design and assembly of DLP bioprinters. This review begins with analyzing unique considerations and specific examples in the hardware, including the resin vat, optical system, and electronics. In the software, the workflow is analyzed, including the parameters to be considered for the control of the bioprinter and the voxelizing/slicing algorithm. In addition, we briefly discuss the material requirements for DLP bioprinting. Then, we provide a section with best practices and maintenance of a do-it-yourself DLP bioprinter. Finally, we highlight the future outlooks of the DLP technology and their critical role in directing the future of bioprinting. The state-of-the-art progress in DLP bioprinter in this review will provide a set of knowledge for innovative DLP bioprinter designs.
Collapse
Affiliation(s)
- Carlos Ezio Garciamendez-Mijares
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Francisco Javier Aguilar
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Pavel Hernandez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Xiao Kuang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Mauricio Gonzalez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Vanessa Ortiz
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Ricardo A. Riesgo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - David S. Rendon Ruiz
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Victoria Abril Manjarrez Rivera
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Juan Carlos Rodriguez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Francisco Lugo Mestre
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Penelope Ceron Castillo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Abraham Perez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Lourdes Monserrat Cruz
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | - Khoon S. Lim
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
4
|
Jiu J, Liu H, Li D, Li J, Liu L, Yang W, Yan L, Li S, Zhang J, Li X, Li JJ, Wang B. 3D bioprinting approaches for spinal cord injury repair. Biofabrication 2024; 16:032003. [PMID: 38569491 DOI: 10.1088/1758-5090/ad3a13] [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: 10/10/2023] [Accepted: 04/03/2024] [Indexed: 04/05/2024]
Abstract
Regenerative healing of spinal cord injury (SCI) poses an ongoing medical challenge by causing persistent neurological impairment and a significant socioeconomic burden. The complexity of spinal cord tissue presents hurdles to successful regeneration following injury, due to the difficulty of forming a biomimetic structure that faithfully replicates native tissue using conventional tissue engineering scaffolds. 3D bioprinting is a rapidly evolving technology with unmatched potential to create 3D biological tissues with complicated and hierarchical structure and composition. With the addition of biological additives such as cells and biomolecules, 3D bioprinting can fabricate preclinical implants, tissue or organ-like constructs, andin vitromodels through precise control over the deposition of biomaterials and other building blocks. This review highlights the characteristics and advantages of 3D bioprinting for scaffold fabrication to enable SCI repair, including bottom-up manufacturing, mechanical customization, and spatial heterogeneity. This review also critically discusses the impact of various fabrication parameters on the efficacy of spinal cord repair using 3D bioprinted scaffolds, including the choice of printing method, scaffold shape, biomaterials, and biological supplements such as cells and growth factors. High-quality preclinical studies are required to accelerate the translation of 3D bioprinting into clinical practice for spinal cord repair. Meanwhile, other technological advances will continue to improve the regenerative capability of bioprinted scaffolds, such as the incorporation of nanoscale biological particles and the development of 4D printing.
Collapse
Affiliation(s)
- Jingwei Jiu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Haifeng Liu
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Dijun Li
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Jiarong Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Lu Liu
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Wenjie Yang
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Lei Yan
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Songyan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jing Zhang
- Department of Emergency Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, People's Republic of China
| | - Xiaoke Li
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
5
|
Huniadi M, Nosálová N, Almášiová V, Horňáková Ľ, Valenčáková A, Hudáková N, Cizkova D. Three-Dimensional Cultivation a Valuable Tool for Modelling Canine Mammary Gland Tumour Behaviour In Vitro. Cells 2024; 13:695. [PMID: 38667310 PMCID: PMC11049302 DOI: 10.3390/cells13080695] [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: 02/29/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cell cultivation has been one of the most popular methods in research for decades. Currently, scientists routinely use two-dimensional (2D) and three-dimensional (3D) cell cultures of commercially available cell lines and primary cultures to study cellular behaviour, responses to stimuli, and interactions with their environment in a controlled laboratory setting. In recent years, 3D cultivation has gained more attention in modern biomedical research, mainly due to its numerous advantages compared to 2D cultures. One of the main goals where 3D culture models are used is the investigation of tumour diseases, in both animals and humans. The ability to simulate the tumour microenvironment and design 3D masses allows us to monitor all the processes that take place in tumour tissue created not only from cell lines but directly from the patient's tumour cells. One of the tumour types for which 3D culture methods are often used in research is the canine mammary gland tumour (CMT). The clinically similar profile of the CMT and breast tumours in humans makes the CMT a suitable model for studying the issue not only in animals but also in women.
Collapse
Affiliation(s)
- Mykhailo Huniadi
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Natália Nosálová
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Viera Almášiová
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Ľubica Horňáková
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Alexandra Valenčáková
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Nikola Hudáková
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Dasa Cizkova
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| |
Collapse
|
6
|
Zamponi M, Mollica PA, Khodour Y, Bjerring JS, Bruno RD, Sachs PC. Combined 3D bioprinting and tissue-specific ECM system reveals the influence of brain matrix on stem cell differentiation. Front Cell Dev Biol 2023; 11:1258993. [PMID: 37928905 PMCID: PMC10623327 DOI: 10.3389/fcell.2023.1258993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
We have previously shown that human and murine breast extracellular matrix (ECM) can significantly impact cellular behavior, including stem cell fate determination. It has been established that tissue-specific extracellular matrix from the central nervous system has the capacity to support neuronal survival. However, the characterization of its influence on stem cell differentiation and its adaptation to robust 3D culture models is underdeveloped. To address these issues, we combined our 3D bioprinter with hydrogels containing porcine brain extracellular matrix (BMX) to test the influence of the extracellular matrix on stem cell differentiation. Our 3D bioprinting system generated reproducible 3D neural structures derived from mouse embryonic stem cells (mESCs). We demonstrate that the addition of BMX preferentially influences 3D bioprinted mESCs towards neural lineages compared to standard basement membrane (Geltrex/Matrigel) hydrogels alone. Furthermore, we demonstrate that we can transplant these 3D bioprinted neural cellular structures into a mouse's cleared mammary fat pad, where they continue to grow into larger neural outgrowths. Finally, we demonstrate that direct injection of human induced pluripotent stem cells (hiPSCS) and neural stem cells (NSCs) suspended in pure BMX formed neural structures in vivo. Combined, these findings describe a unique system for studying brain ECM/stem cell interactions and demonstrate that BMX can direct pluripotent stem cells to differentiate down a neural cellular lineage without any additional specific differentiation stimuli.
Collapse
Affiliation(s)
- Martina Zamponi
- School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, VA, United States
| | - Peter A. Mollica
- School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, VA, United States
| | - Yara Khodour
- School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, VA, United States
| | - Julie S. Bjerring
- School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, VA, United States
| | - Robert D. Bruno
- School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, VA, United States
| | - Patrick C. Sachs
- School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, VA, United States
| |
Collapse
|
7
|
Yang Q, Li M, Yang X, Xiao Z, Tong X, Tuerdi A, Li S, Lei L. Flourishing tumor organoids: History, emerging technology, and application. Bioeng Transl Med 2023; 8:e10559. [PMID: 37693042 PMCID: PMC10487342 DOI: 10.1002/btm2.10559] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/16/2023] [Accepted: 05/25/2023] [Indexed: 09/12/2023] Open
Abstract
Malignant tumors are one of the leading causes of death which impose an increasingly heavy burden on all countries. Therefore, the establishment of research models that closely resemble original tumor characteristics is crucial to further understanding the mechanisms of malignant tumor development, developing safer and more effective drugs, and formulating personalized treatment plans. Recently, organoids have been widely used in tumor research owing to their advantages including preserving the structure, heterogeneity, and cellular functions of the original tumor, together with the ease of manipulation. This review describes the history and characteristics of tumor organoids and the synergistic combination of three-dimensional (3D) culture approaches for tumor organoids with emerging technologies, including tissue-engineered cell scaffolds, microfluidic devices, 3D bioprinting, rotating wall vessels, and clustered regularly interspaced short palindromic repeats-CRISPR-associated protein 9 (CRISPR-Cas9). Additionally, the progress in research and the applications in basic and clinical research of tumor organoid models are summarized. This includes studies of the mechanism of tumor development, drug development and screening, precision medicine, immunotherapy, and simulation of the tumor microenvironment. Finally, the existing shortcomings of tumor organoids and possible future directions are discussed.
Collapse
Affiliation(s)
- Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zian Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ayinuer Tuerdi
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| |
Collapse
|
8
|
D'Antoni C, Mautone L, Sanchini C, Tondo L, Grassmann G, Cidonio G, Bezzi P, Cordella F, Di Angelantonio S. Unlocking Neural Function with 3D In Vitro Models: A Technical Review of Self-Assembled, Guided, and Bioprinted Brain Organoids and Their Applications in the Study of Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2023; 24:10762. [PMID: 37445940 DOI: 10.3390/ijms241310762] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Understanding the complexities of the human brain and its associated disorders poses a significant challenge in neuroscience. Traditional research methods have limitations in replicating its intricacies, necessitating the development of in vitro models that can simulate its structure and function. Three-dimensional in vitro models, including organoids, cerebral organoids, bioprinted brain models, and functionalized brain organoids, offer promising platforms for studying human brain development, physiology, and disease. These models accurately replicate key aspects of human brain anatomy, gene expression, and cellular behavior, enabling drug discovery and toxicology studies while providing insights into human-specific phenomena not easily studied in animal models. The use of human-induced pluripotent stem cells has revolutionized the generation of 3D brain structures, with various techniques developed to generate specific brain regions. These advancements facilitate the study of brain structure development and function, overcoming previous limitations due to the scarcity of human brain samples. This technical review provides an overview of current 3D in vitro models of the human cortex, their development, characterization, and limitations, and explores the state of the art and future directions in the field, with a specific focus on their applications in studying neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Chiara D'Antoni
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Lorenza Mautone
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Caterina Sanchini
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Lucrezia Tondo
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Greta Grassmann
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, 00185 Rome, Italy
| | - Gianluca Cidonio
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Paola Bezzi
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Department of Fundamental Neurosciences, University of Lausanne, 1011 Lausanne, Switzerland
| | - Federica Cordella
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- D-Tails s.r.l., 00165 Rome, Italy
| |
Collapse
|
9
|
Pang S, Wu D, Gurlo A, Kurreck J, Hanaor DAH. Additive manufacturing and performance of bioceramic scaffolds with different hollow strut geometries. Biofabrication 2023; 15. [PMID: 36645921 DOI: 10.1088/1758-5090/acb387] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/16/2023] [Indexed: 01/18/2023]
Abstract
Additively manufactured hollow-strut bioceramic scaffolds present a promising strategy towards enhanced performance in patient-tailored bone tissue engineering. The channels in such scaffolds offer pathways for nutrient and cell transport and facilitate effective osseointegration and vascularization. In this study, we report an approach for the slurry based additive manufacturing of modified diopside bioceramics that enables the production of hollow-strut scaffolds with diverse cross-sectional forms, distinguished by different configurations of channel and strut geometries. The prepared scaffolds exhibit levels of porosity and mechanical strength that are well suited for osteoporotic bone repair. Mechanical characterization in orthogonal orientations revealed that a square outer cross-section for hollow struts in woodpile scaffolds gives rise to levels of compressive strength that are higher than those of conventional solid cylindrical strut scaffolds despite a significantly lower density. Finite element analysis confirms that this improved strength arises from lower stress concentration in such geometries. It was shown that hollow struts in bioceramic scaffolds dramatically increase cell attachment and proliferation, potentially promoting new bone tissue formation within the scaffold channel. This work provides an easily controlled method for the extrusion-based 3D printing of hollow strut scaffolds. We show here how the production of hollow struts with controllable geometry can serve to enhance both the functional and mechanical performance of porous structures, with particular relevance for bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Shumin Pang
- Technische Universität Berlin, Chair of Advanced Ceramic Materials, Straße des 17. Juni 135, 10623 Berlin, Germany
| | - Dongwei Wu
- Technische Universität Berlin, Chair of Applied Biochemistry, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Aleksander Gurlo
- Technische Universität Berlin, Chair of Advanced Ceramic Materials, Straße des 17. Juni 135, 10623 Berlin, Germany
| | - Jens Kurreck
- Technische Universität Berlin, Chair of Applied Biochemistry, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Dorian A H Hanaor
- Technische Universität Berlin, Chair of Advanced Ceramic Materials, Straße des 17. Juni 135, 10623 Berlin, Germany
| |
Collapse
|
10
|
Mazzaglia C, Sheng Y, Rodrigues LN, Lei IM, Shields JD, Huang YYS. Deployable extrusion bioprinting of compartmental tumoroids with cancer associated fibroblasts for immune cell interactions. Biofabrication 2023; 15:025005. [PMID: 36626838 DOI: 10.1088/1758-5090/acb1db] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/10/2023] [Indexed: 01/11/2023]
Abstract
Realizing the translational impacts of three-dimensional (3D) bioprinting for cancer research necessitates innovation in bioprinting workflows which integrate affordability, user-friendliness, and biological relevance. Herein, we demonstrate 'BioArm', a simple, yet highly effective extrusion bioprinting platform, which can be folded into a carry-on pack, and rapidly deployed between bio-facilities. BioArm enabled the reconstruction of compartmental tumoroids with cancer-associated fibroblasts (CAFs), forming the shell of each tumoroid. The 3D printed core-shell tumoroids showedde novosynthesized extracellular matrices, and enhanced cellular proliferation compared to the tumour alone 3D printed spheroid culture. Further, thein vivophenotypes of CAFs normally lost after conventional 2D co-culture re-emerged in the bioprinted model. Embedding the 3D printed tumoroids in an immune cell-laden collagen matrix permitted tracking of the interaction between immune cells and tumoroids, and subsequent simulated immunotherapy treatments. Our deployable extrusion bioprinting workflow could significantly widen the accessibility of 3D bioprinting for replicating multi-compartmental architectures of tumour microenvironment, and for developing strategies in cancer drug testing in the future.
Collapse
Affiliation(s)
| | - Yaqi Sheng
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | | | - Iek Man Lei
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Jacqueline D Shields
- MRC Cancer Unit, University of Cambridge, Cambridge, United Kingdom
- Comprehensive Cancer Centre, King's College London, Great Maze Pond, London, United Kingdom
| | - Yan Yan Shery Huang
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Loukelis K, Helal ZA, Mikos AG, Chatzinikolaidou M. Nanocomposite Bioprinting for Tissue Engineering Applications. Gels 2023; 9:103. [PMID: 36826273 PMCID: PMC9956920 DOI: 10.3390/gels9020103] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Bioprinting aims to provide new avenues for regenerating damaged human tissues through the controlled printing of live cells and biocompatible materials that can function therapeutically. Polymeric hydrogels are commonly investigated ink materials for 3D and 4D bioprinting applications, as they can contain intrinsic properties relative to those of the native tissue extracellular matrix and can be printed to produce scaffolds of hierarchical organization. The incorporation of nanoscale material additives, such as nanoparticles, to the bulk of inks, has allowed for significant tunability of the mechanical, biological, structural, and physicochemical material properties during and after printing. The modulatory and biological effects of nanoparticles as bioink additives can derive from their shape, size, surface chemistry, concentration, and/or material source, making many configurations of nanoparticle additives of high interest to be thoroughly investigated for the improved design of bioactive tissue engineering constructs. This paper aims to review the incorporation of nanoparticles, as well as other nanoscale additive materials, to printable bioinks for tissue engineering applications, specifically bone, cartilage, dental, and cardiovascular tissues. An overview of the various bioinks and their classifications will be discussed with emphasis on cellular and mechanical material interactions, as well the various bioink formulation methodologies for 3D and 4D bioprinting techniques. The current advances and limitations within the field will be highlighted.
Collapse
Affiliation(s)
- Konstantinos Loukelis
- Department of Materials Science and Technology, University of Crete, 70013 Heraklion, Greece
| | - Zina A. Helal
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Antonios G. Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, 70013 Heraklion, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FO.R.T.H), 70013 Heraklion, Greece
| |
Collapse
|
12
|
Fathi S, Lei IM, Cao Y, Huang YYS. Microcapillary cell extrusion deposition with picolitre dispensing resolution. Biodes Manuf 2023; 6:1-11. [PMID: 36644556 PMCID: PMC9829649 DOI: 10.1007/s42242-022-00205-3] [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: 11/25/2021] [Accepted: 06/15/2022] [Indexed: 01/18/2023]
Abstract
Extrusion-based cell deposition has become a prominent technique for expanding bioprinting applications. However, the associated print resolution in the order of nanolitre or above has been a limiting factor. The demand for improving print resolution towards the scale of a single cell has driven the development of precision nozzle extrusion, although the benefits gained remain ambiguous. Here, aided by in situ imaging, we investigated the dynamics of cell organisation through an extrusion-based microcapillary tip with picolitre precision through in-air or immersion deposition. The microcapillary extrusion setup, termed 'Picodis', was demonstrated by generating droplets of colouring inks immersed in an immiscible medium. Next, using 3T3 fibroblast cells as an experimental model, we demonstrated the deposition of cell suspension, and pre-aggregated cell pellets. Then, the dynamic organisation of cells within the microcapillary tip was described, along with cell ejection and deposition upon exiting the tip opening. The vision-assisted approach revealed that when dispersed in a culture medium, the movements of cells were distinctive based on the flow profiles and were purely driven by laminar fluid flow within a narrow tip. The primary process limitations were cell sedimentation, aggregation and compaction, along with trapped air bubbles. The use of picolitre-level resolution microcapillary extrusion, although it provides some level of control for a small number of cells, does not necessarily offer a reliable method when a specified number of cells are required. Our study provides insights into the process limitations of high-resolution cell ink extrusion, which may be useful for optimising biofabrication processes of cell-laden constructs for biomedical research. Supplementary information The online version contains supplementary material available at 10.1007/s42242-022-00205-3.
Collapse
Affiliation(s)
- Saeed Fathi
- Department of Engineering, University of Cambridge, Cambridge, UK ,The Nanoscience Centre, University of Cambridge, Cambridge, UK
| | - Iek Man Lei
- Department of Engineering, University of Cambridge, Cambridge, UK ,The Nanoscience Centre, University of Cambridge, Cambridge, UK
| | - Yang Cao
- Department of Engineering, University of Cambridge, Cambridge, UK ,The Nanoscience Centre, University of Cambridge, Cambridge, UK
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Cambridge, UK ,The Nanoscience Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
13
|
Can 3D bioprinting solve the mystery of senescence in cancer therapy? Ageing Res Rev 2022; 81:101732. [PMID: 36100069 DOI: 10.1016/j.arr.2022.101732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/30/2022] [Accepted: 09/08/2022] [Indexed: 01/31/2023]
Abstract
Tumor dormancy leading to cancer relapse is still a poorly understood mechanism. Several cell states such as quiescence and diapause can explain the persistence of tumor cells in a dormant state, but the potential role of tumor cell senescence has been met with hesitance given the historical understanding of the senescent growth arrest as irreversible. However, recent evidence has suggested that senescence might contribute to dormancy and relapse, although its exact role is not fully developed. This limited understanding is largely due to the paucity of reliable study models. The current 2D cell modeling is overly simplistic and lacks the appropriate representation of the interactions between tumor cells (senescent or non-senescent) and the other cell types within the tumor microenvironment (TME), as well as with the extracellular matrix (ECM). 3D cell culture models, including 3D bioprinting techniques, offer a promising approach to better recapitulate the native cancer microenvironment and would significantly improve our understanding of cancer biology and cellular response to treatment, particularly Therapy-Induced Senescence (TIS), and its contribution to tumor dormancy and cancer recurrence. Fabricating a novel 3D bioprinted model offers excellent opportunities to investigate both the role of TIS in tumor dormancy and the utility of senolytics (drugs that selectively eliminate senescent cells) in targeting dormant cancer cells and mitigating the risk for resurgence. In this review, we discuss literature on the possible contribution of TIS in tumor dormancy, provide examples on the current 3D models of senescence, and propose a novel 3D model to investigate the ultimate role of TIS in mediating overall response to therapy.
Collapse
|
14
|
A hackable, multi-functional, and modular extrusion 3D printer for soft materials. Sci Rep 2022; 12:12294. [PMID: 35853916 PMCID: PMC9296631 DOI: 10.1038/s41598-022-16008-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/04/2022] [Indexed: 12/26/2022] Open
Abstract
Three-dimensional (3D) printing has emerged as a powerful tool for material, food, and life science research and development, where the technology’s democratization necessitates the advancement of open-source platforms. Herein, we developed a hackable, multi-functional, and modular extrusion 3D printer for soft materials, nicknamed Printer.HM. Multi-printhead modules are established based on a robotic arm for heterogeneous construct creation, where ink printability can be tuned by accessories such as heating and UV modules. Software associated with Printer.HM were designed to accept geometry inputs including computer-aided design models, coordinates, equations, and pictures, to create prints of distinct characteristics. Printer.HM could further perform versatile operations, such as liquid dispensing, non-planar printing, and pick-and-place of meso-objects. By ‘mix-and-match’ software and hardware settings, Printer.HM demonstrated printing of pH-responsive soft actuators, plant-based functional hydrogels, and organ macro-anatomical models. Integrating affordability and open design, Printer.HM is envisaged to democratize 3D printing for soft, biological, and sustainable material architectures.
Collapse
|
15
|
Abstract
Three-dimensional printing is a still-emerging technology with high impact for the medical community, particularly in the development of tissues for the clinic. Many types of printers are under development, including extrusion, droplet, melt, and light-curing technologies. Herein we discuss the various types of 3D printers and their strengths and weaknesses concerning tissue engineering. Despite the advantages of 3D printing, challenges remain in the development of large, clinically relevant tissues. Advancements in bioink development, printer technology, tissue vascularization, and cellular sourcing/expansion are discussed, alongside future opportunities for the field. Trends regarding in situ printing, personalized medicine, and whole organ development are highlighted. Expected final online publication date for the Annual Review of Chemical and Biomolecular Engineering, Volume 13 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kelsey Willson
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA;
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA;
| |
Collapse
|
16
|
Shukla AK, Gao G, Kim BS. Applications of 3D Bioprinting Technology in Induced Pluripotent Stem Cells-Based Tissue Engineering. MICROMACHINES 2022; 13:155. [PMID: 35208280 PMCID: PMC8876961 DOI: 10.3390/mi13020155] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are essentially produced by the genetic reprogramming of adult cells. Moreover, iPSC technology prevents the genetic manipulation of embryos. Hence, with the ensured element of safety, they rarely cause ethical concerns when utilized in tissue engineering. Several cumulative outcomes have demonstrated the functional superiority and potency of iPSCs in advanced regenerative medicine. Recently, an emerging trend in 3D bioprinting technology has been a more comprehensive approach to iPSC-based tissue engineering. The principal aim of this review is to provide an understanding of the applications of 3D bioprinting in iPSC-based tissue engineering. This review discusses the generation of iPSCs based on their distinct purpose, divided into two categories: (1) undifferentiated iPSCs applied with 3D bioprinting; (2) differentiated iPSCs applied with 3D bioprinting. Their significant potential is analyzed. Lastly, various applications for engineering tissues and organs have been introduced and discussed in detail.
Collapse
Affiliation(s)
- Arvind Kumar Shukla
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Korea;
| | - Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China
- Department of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Korea;
| |
Collapse
|
17
|
Ravanbakhsh H, Karamzadeh V, Bao G, Mongeau L, Juncker D, Zhang YS. Emerging Technologies in Multi-Material Bioprinting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2104730. [PMID: 34596923 PMCID: PMC8971140 DOI: 10.1002/adma.202104730] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/10/2021] [Indexed: 05/09/2023]
Abstract
Bioprinting, within the emerging field of biofabrication, aims at the fabrication of functional biomimetic constructs. Different 3D bioprinting techniques have been adapted to bioprint cell-laden bioinks. However, single-material bioprinting techniques oftentimes fail to reproduce the complex compositions and diversity of native tissues. Multi-material bioprinting as an emerging approach enables the fabrication of heterogeneous multi-cellular constructs that replicate their host microenvironments better than single-material approaches. Here, bioprinting modalities are reviewed, their being adapted to multi-material bioprinting is discussed, and their advantages and challenges, encompassing both custom-designed and commercially available technologies are analyzed. A perspective of how multi-material bioprinting opens up new opportunities for tissue engineering, tissue model engineering, therapeutics development, and personalized medicine is offered.
Collapse
Affiliation(s)
- Hossein Ravanbakhsh
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, McGill University, Montreal, QC, H3A0C3, Canada
| | - Vahid Karamzadeh
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3A0G1, Canada
| | - Guangyu Bao
- Department of Mechanical Engineering, McGill University, Montreal, QC, H3A0C3, Canada
| | - Luc Mongeau
- Department of Mechanical Engineering, McGill University, Montreal, QC, H3A0C3, Canada
| | - David Juncker
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3A0G1, Canada
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| |
Collapse
|
18
|
Tarassoli SP, Jessop ZM, Jovic T, Hawkins K, Whitaker IS. Candidate Bioinks for Extrusion 3D Bioprinting-A Systematic Review of the Literature. Front Bioeng Biotechnol 2021; 9:616753. [PMID: 34722473 PMCID: PMC8548422 DOI: 10.3389/fbioe.2021.616753] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/19/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose: Bioprinting is becoming an increasingly popular platform technology for engineering a variety of tissue types. Our aim was to identify biomaterials that have been found to be suitable for extrusion 3D bioprinting, outline their biomechanical properties and biocompatibility towards their application for bioprinting specific tissue types. This systematic review provides an in-depth overview of current biomaterials suitable for extrusion to aid bioink selection for specific research purposes and facilitate design of novel tailored bioinks. Methods: A systematic search was performed on EMBASE, PubMed, Scopus and Web of Science databases according to the PRISMA guidelines. References of relevant articles, between December 2006 to January 2018, on candidate bioinks used in extrusion 3D bioprinting were reviewed by two independent investigators against standardised inclusion and exclusion criteria. Data was extracted on bioprinter brand and model, printing technique and specifications (speed and resolution), bioink material and class of mechanical assessment, cell type, viability, and target tissue. Also noted were authors, study design (in vitro/in vivo), study duration and year of publication. Results: A total of 9,720 studies were identified, 123 of which met inclusion criteria, consisting of a total of 58 reports using natural biomaterials, 26 using synthetic biomaterials and 39 using a combination of biomaterials as bioinks. Alginate (n = 50) and PCL (n = 33) were the most commonly used bioinks, followed by gelatin (n = 18) and methacrylated gelatin (GelMA) (n = 16). Pneumatic extrusion bioprinting techniques were the most common (n = 78), followed by piston (n = 28). The majority of studies focus on the target tissue, most commonly bone and cartilage, and investigate only one bioink rather than assessing a range to identify those with the most promising printability and biocompatibility characteristics. The Bioscaffolder (GeSiM, Germany), 3D Discovery (regenHU, Switzerland), and Bioplotter (EnvisionTEC, Germany) were the most commonly used commercial bioprinters (n = 35 in total), but groups most often opted to create their own in-house devices (n = 20). Many studies also failed to specify whether the mechanical data reflected pre-, during or post-printing, pre- or post-crosslinking and with or without cells. Conclusions: Despite the continued increase in the variety of biocompatible synthetic materials available, there has been a shift change towards using natural rather than synthetic bioinks for extrusion bioprinting, dominated by alginate either alone or in combination with other biomaterials. On qualitative analysis, no link was demonstrated between the type of bioink or extrusion technique and the target tissue, indicating that bioprinting research is in its infancy with no established tissue specific bioinks or bioprinting techniques. Further research is needed on side-by-side characterisation of bioinks with standardisation of the type and timing of biomechanical assessment.
Collapse
Affiliation(s)
- Sam P Tarassoli
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom
| | - Zita M Jessop
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom.,The Welsh Centre for Burns & Plastic Surgery, Morriston Hospital, Swansea, United Kingdom
| | - Thomas Jovic
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom.,The Welsh Centre for Burns & Plastic Surgery, Morriston Hospital, Swansea, United Kingdom
| | - Karl Hawkins
- Centre for NanoHealth, Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom
| | - Iain S Whitaker
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom.,The Welsh Centre for Burns & Plastic Surgery, Morriston Hospital, Swansea, United Kingdom
| |
Collapse
|
19
|
Xu ZY, Huang JJ, Liu Y, Zhao Y, Wu XW, Ren JA. Current knowledge on the multiform reconstitution of intestinal stem cell niche. World J Stem Cells 2021; 13:1564-1579. [PMID: 34786158 PMCID: PMC8567451 DOI: 10.4252/wjsc.v13.i10.1564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/02/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
The development of “mini-guts” organoid originates from the identification of Lgr5+ intestinal stem cells (ISCs) and circumambient signalings within their specific niche at the crypt bottom. These in vitro self-renewing “mini-guts”, also named enteroids or colonoids, undergo perpetual proliferation and regulated differentiation, which results in a high-performance, self-assembling and physiological organoid platform in diverse areas of intestinal research and therapy. The triumphant reconstitution of ISC niche in vitro also relies on Matrigel, a heterogeneous sarcoma extract. Despite the promising prospect of organoids research, their expanding applications are hampered by the canonical culture pattern, which reveals limitations such as inaccessible lumen, confine scale, batch to batch variation and low reproducibility. The tumor-origin of Matrigel also raises biosafety concerns in clinical treatment. However, the convergence of breakthroughs in cellular biology and bioengineering contribute to multiform reconstitution of the ISC niche. Herein, we review the recent advances in the microfabrication of intestinal organoids on hydrogel systems.
Collapse
Affiliation(s)
- Zi-Yan Xu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Jin-Jian Huang
- Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Ye Liu
- Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, Nanjing 210019, Jiangsu Province, China
| | - Xiu-Wen Wu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Jian-An Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| |
Collapse
|
20
|
Development and Application of 3D Bioprinted Scaffolds Supporting Induced Pluripotent Stem Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:4910816. [PMID: 34552987 PMCID: PMC8452409 DOI: 10.1155/2021/4910816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Three-dimensional (3D) bioprinting is a revolutionary technology that replicates 3D functional living tissue scaffolds in vitro by controlling the layer-by-layer deposition of biomaterials and enables highly precise positioning of cells. With the development of this technology, more advanced research on the mechanisms of tissue morphogenesis, clinical drug screening, and organ regeneration may be pursued. Because of their self-renewal characteristics and multidirectional differentiation potential, induced pluripotent stem cells (iPSCs) have outstanding advantages in stem cell research and applications. In this review, we discuss the advantages of different bioinks containing human iPSCs that are fabricated by using 3D bioprinting. In particular, we focus on the ability of these bioinks to support iPSCs and promote their proliferation and differentiation. In addition, we summarize the applications of 3D bioprinting with iPSC-containing bioinks and put forward new views on the current research status.
Collapse
|
21
|
Jin Y, Cho SW. Bioengineering platforms for cell therapeutics derived from pluripotent and direct reprogramming. APL Bioeng 2021; 5:031501. [PMID: 34258498 PMCID: PMC8263070 DOI: 10.1063/5.0040621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
Pluripotent and direct reprogramming technologies hold great potential for tissue repair and restoration of tissue and organ function. The implementation of induced pluripotent stem cells and directly reprogrammed cells in biomedical research has resulted in a significant leap forward in the highly promising area of regenerative medicine. While these therapeutic strategies are promising, there are several obstacles to overcome prior to the introduction of these therapies into clinical settings. Bioengineering technologies, such as biomaterials, bioprinting, microfluidic devices, and biostimulatory systems, can enhance cell viability, differentiation, and function, in turn the efficacy of cell therapeutics generated via pluripotent and direct reprogramming. Therefore, cellular reprogramming technologies, in combination with tissue-engineering platforms, are poised to overcome current bottlenecks associated with cell-based therapies and create new ways of producing engineered tissue substitutes.
Collapse
Affiliation(s)
- Yoonhee Jin
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | | |
Collapse
|
22
|
Halim A, Qu KY, Zhang XF, Huang NP. Recent Advances in the Application of Two-Dimensional Nanomaterials for Neural Tissue Engineering and Regeneration. ACS Biomater Sci Eng 2021; 7:3503-3529. [PMID: 34291638 DOI: 10.1021/acsbiomaterials.1c00490] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The complexity of the nervous system structure and function, and its slow regeneration rate, makes it more difficult to treat compared to other tissues in the human body when an injury occurs. Moreover, the current therapeutic approaches including the use of autografts, allografts, and pharmacological agents have several drawbacks and can not fully restore nervous system injuries. Recently, nanotechnology and tissue engineering approaches have attracted many researchers to guide tissue regeneration in an effective manner. Owing to their remarkable physicochemical and biological properties, two-dimensional (2D) nanomaterials have been extensively studied in the tissue engineering and regenerative medicine field. The great conductivity of these materials makes them a promising candidate for the development of novel scaffolds for neural tissue engineering application. Moreover, the high loading capacity of 2D nanomaterials also has attracted many researchers to utilize them as a drug/gene delivery method to treat various devastating nervous system disorders. This review will first introduce the fundamental physicochemical properties of 2D nanomaterials used in biomedicine and the supporting biological properties of 2D nanomaterials for inducing neuroregeneration, including their biocompatibility on neural cells, the ability to promote the neural differentiation of stem cells, and their immunomodulatory properties which are beneficial for alleviating chronic inflammation at the site of the nervous system injury. It also discusses various types of 2D nanomaterials-based scaffolds for neural tissue engineering applications. Then, the latest progress on the use of 2D nanomaterials for nervous system disorder treatment is summarized. Finally, a discussion of the challenges and prospects of 2D nanomaterials-based applications in neural tissue engineering is provided.
Collapse
Affiliation(s)
- Alexander Halim
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Kai-Yun Qu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Xiao-Feng Zhang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Ning-Ping Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| |
Collapse
|
23
|
Tong A, Pham QL, Abatemarco P, Mathew A, Gupta D, Iyer S, Voronov R. Review of Low-Cost 3D Bioprinters: State of the Market and Observed Future Trends. SLAS Technol 2021; 26:333-366. [PMID: 34137286 DOI: 10.1177/24726303211020297] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three-dimensional (3D) bioprinting has become mainstream for precise and repeatable high-throughput fabrication of complex cell cultures and tissue constructs in drug testing and regenerative medicine, food products, dental and medical implants, biosensors, and so forth. Due to this tremendous growth in demand, an overwhelming amount of hardware manufacturers have recently flooded the market with different types of low-cost bioprinter models-a price segment that is most affordable to typical-sized laboratories. These machines range in sophistication, type of the underlying printing technology, and possible add-ons/features, which makes the selection process rather daunting (especially for a nonexpert customer). Yet, the review articles available in the literature mostly focus on the technical aspects of the printer technologies under development, as opposed to explaining the differences in what is already on the market. In contrast, this paper provides a snapshot of the fast-evolving low-cost bioprinter niche, as well as reputation profiles (relevant to delivery time, part quality, adherence to specifications, warranty, maintenance, etc.) of the companies selling these machines. Specifically, models spanning three dominant technologies-microextrusion, droplet-based/inkjet, and light-based/crosslinking-are reviewed. Additionally, representative examples of high-end competitors (including up-and-coming microfluidics-based bioprinters) are discussed to highlight their major differences and advantages relative to the low-cost models. Finally, forecasts are made based on the trends observed during this survey, as to the anticipated trickling down of the high-end technologies to the low-cost printers. Overall, this paper provides insight for guiding buyers on a limited budget toward making informed purchasing decisions in this fast-paced market.
Collapse
Affiliation(s)
- Anh Tong
- The Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology Newark College of Engineering, Newark, NJ, USA
| | - Quang Long Pham
- The Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology Newark College of Engineering, Newark, NJ, USA
| | - Paul Abatemarco
- The Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology Newark College of Engineering, Newark, NJ, USA
| | - Austin Mathew
- Department of Biomedical Engineering, New Jersey Institute of Technology Newark College of Engineering, Newark, NJ, USA
| | - Dhruv Gupta
- The Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology Newark College of Engineering, Newark, NJ, USA
| | - Siddharth Iyer
- The Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology Newark College of Engineering, Newark, NJ, USA
| | - Roman Voronov
- The Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology Newark College of Engineering, Newark, NJ, USA.,Department of Biomedical Engineering, New Jersey Institute of Technology Newark College of Engineering, Newark, NJ, USA
| |
Collapse
|
24
|
Gómez-Blanco JC, Galván-Chacón V, Patrocinio D, Matamoros M, Sánchez-Ortega ÁJ, Marcos AC, Duarte-León M, Marinaro F, Pagador JB, Sánchez-Margallo FM. Improving Cell Viability and Velocity in μ-Extrusion Bioprinting with a Novel Pre-Incubator Bioprinter and a Standard FDM 3D Printing Nozzle. MATERIALS (BASEL, SWITZERLAND) 2021; 14:3100. [PMID: 34198815 PMCID: PMC8201198 DOI: 10.3390/ma14113100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022]
Abstract
Bioprinting is a promising emerging technology. It has been widely studied by the scientific community for the possibility to create transplantable artificial tissues, with minimal risk to the patient. Although the biomaterials and cells to be used are being carefully studied, there is still a long way to go before a bioprinter can easily and quickly produce printings without harmful effects on the cells. In this sense, we have developed a new μ-extrusion bioprinter formed by an Atom Proton 3D printer, an atmospheric enclosure and a new extrusion-head capable to increment usual printing velocity. Hence, this work has two main objectives. First, to experimentally study the accuracy and precision. Secondly, to study the influence of flow rates on cellular viability using this novel μ-extrusion bioprinter in combination with a standard FDM 3D printing nozzle. Our results show an X, Y and Z axis movement accuracy under 17 μm with a precision around 12 μm while the extruder values are under 5 and 7 μm, respectively. Additionally, the cell viability obtained from different volumetric flow tests varies from 70 to 90%. So, the proposed bioprinter and nozzle can control the atmospheric conditions and increase the volumetric flow speeding up the bioprinting process without compromising the cell viability.
Collapse
Affiliation(s)
- Juan C. Gómez-Blanco
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (V.G.-C.); (D.P.); (M.D.-L.); (F.M.); (F.M.S.-M.)
| | - Victor Galván-Chacón
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (V.G.-C.); (D.P.); (M.D.-L.); (F.M.); (F.M.S.-M.)
| | - David Patrocinio
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (V.G.-C.); (D.P.); (M.D.-L.); (F.M.); (F.M.S.-M.)
| | - Manuel Matamoros
- School of Industrial Engineering, University of Extremadura, 06006 Badajoz, Spain; (M.M.); (Á.J.S.-O.); (A.C.M.)
| | - Álvaro J. Sánchez-Ortega
- School of Industrial Engineering, University of Extremadura, 06006 Badajoz, Spain; (M.M.); (Á.J.S.-O.); (A.C.M.)
| | - Alfonso C. Marcos
- School of Industrial Engineering, University of Extremadura, 06006 Badajoz, Spain; (M.M.); (Á.J.S.-O.); (A.C.M.)
| | - María Duarte-León
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (V.G.-C.); (D.P.); (M.D.-L.); (F.M.); (F.M.S.-M.)
| | - Federica Marinaro
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (V.G.-C.); (D.P.); (M.D.-L.); (F.M.); (F.M.S.-M.)
| | - José B. Pagador
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (V.G.-C.); (D.P.); (M.D.-L.); (F.M.); (F.M.S.-M.)
| | - Francisco M. Sánchez-Margallo
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (V.G.-C.); (D.P.); (M.D.-L.); (F.M.); (F.M.S.-M.)
| |
Collapse
|
25
|
Carvalho V, Gonçalves I, Lage T, Rodrigues RO, Minas G, Teixeira SFCF, Moita AS, Hori T, Kaji H, Lima RA. 3D Printing Techniques and Their Applications to Organ-on-a-Chip Platforms: A Systematic Review. SENSORS (BASEL, SWITZERLAND) 2021; 21:3304. [PMID: 34068811 PMCID: PMC8126238 DOI: 10.3390/s21093304] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/14/2021] [Accepted: 05/06/2021] [Indexed: 02/01/2023]
Abstract
Three-dimensional (3D) in vitro models, such as organ-on-a-chip platforms, are an emerging and effective technology that allows the replication of the function of tissues and organs, bridging the gap amid the conventional models based on planar cell cultures or animals and the complex human system. Hence, they have been increasingly used for biomedical research, such as drug discovery and personalized healthcare. A promising strategy for their fabrication is 3D printing, a layer-by-layer fabrication process that allows the construction of complex 3D structures. In contrast, 3D bioprinting, an evolving biofabrication method, focuses on the accurate deposition of hydrogel bioinks loaded with cells to construct tissue-engineered structures. The purpose of the present work is to conduct a systematic review (SR) of the published literature, according to the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses, providing a source of information on the evolution of organ-on-a-chip platforms obtained resorting to 3D printing and bioprinting techniques. In the literature search, PubMed, Scopus, and ScienceDirect databases were used, and two authors independently performed the search, study selection, and data extraction. The goal of this SR is to highlight the importance and advantages of using 3D printing techniques in obtaining organ-on-a-chip platforms, and also to identify potential gaps and future perspectives in this research field. Additionally, challenges in integrating sensors in organs-on-chip platforms are briefly investigated and discussed.
Collapse
Affiliation(s)
- Violeta Carvalho
- MEtRICs, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
| | - Inês Gonçalves
- MEtRICs, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
| | - Teresa Lage
- Center for MicroElectromechanical Systems (CMEMS-UMinho), Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal; (T.L.); (R.O.R.); (G.M.)
| | - Raquel O. Rodrigues
- Center for MicroElectromechanical Systems (CMEMS-UMinho), Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal; (T.L.); (R.O.R.); (G.M.)
| | - Graça Minas
- Center for MicroElectromechanical Systems (CMEMS-UMinho), Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal; (T.L.); (R.O.R.); (G.M.)
| | | | - Ana S. Moita
- IN+, Center for Innovation, Technology and Policy Research, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal;
- CINAMIL, Department of Exact Sciences and Engineering, Portuguese Military Academy, R. Gomes Freire 203, 1169-203 Lisboa, Portugal
| | - Takeshi Hori
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan; (T.H.); (H.K.)
| | - Hirokazu Kaji
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan; (T.H.); (H.K.)
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, Sendai 980-8579, Japan
| | - Rui A. Lima
- MEtRICs, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
- CEFT, Faculty of Engineering of the University of Porto (FEUP), R. Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
26
|
Sayed IM, El-Hafeez AAA, Maity PP, Das S, Ghosh P. Modeling colorectal cancers using multidimensional organoids. Adv Cancer Res 2021; 151:345-383. [PMID: 34148617 PMCID: PMC8221168 DOI: 10.1016/bs.acr.2021.02.005] [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] [Indexed: 10/21/2022]
Abstract
Organoids have revolutionized cancer research as highly adaptable models that enable an array of experimental techniques to interrogate tissue morphology and function. Because they preserve the genetic, phenotypic, and behavioral traits of their source tissue, organoids have gained traction as the most relevant models for drug discovery, tracking therapeutic response and for personalized medicine. As organoids are indisputably becoming a mainstay of cancer research, this review specifically addresses how colon-derived organoids can be perfected as multidimensional, scalable, reproducible models of healthy, pre-neoplastic and neoplastic conditions of the colon and for use in high-throughput "Phase-0" human clinical trials-in-a-dish.
Collapse
Affiliation(s)
- Ibrahim M Sayed
- Department of Pathology, University of California, San Diego, CA, United States
| | - Amer Ali Abd El-Hafeez
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, United States
| | - Priti P Maity
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, United States
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA, United States; Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA, United States; HUMANOID Center of Research Excellence (CoRE), University of California, San Diego, CA, United States.
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, United States; Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA, United States; Department of Medicine, University of California, San Diego, CA, United States; Veterans Affairs Medical Center, San Diego, CA, United States; HUMANOID Center of Research Excellence (CoRE), University of California, San Diego, CA, United States.
| |
Collapse
|
27
|
Sun M, Liu A, Yang X, Gong J, Yu M, Yao X, Wang H, He Y. 3D Cell Culture—Can It Be As Popular as 2D Cell Culture? ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Miao Sun
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - An Liu
- Department of Orthopaedic Surgery Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou 310000 China
| | - Xiaofu Yang
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Jiaxing Gong
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Mengfei Yu
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Xinhua Yao
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
| | - Huiming Wang
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Yong He
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
- State Key Laboratory of Fluid Power and Mechatronic Systems School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
| |
Collapse
|
28
|
Ruiz-Alonso S, Villate-Beitia I, Gallego I, Lafuente-Merchan M, Puras G, Saenz-del-Burgo L, Pedraz JL. Current Insights Into 3D Bioprinting: An Advanced Approach for Eye Tissue Regeneration. Pharmaceutics 2021; 13:pharmaceutics13030308. [PMID: 33653003 PMCID: PMC7996883 DOI: 10.3390/pharmaceutics13030308] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/19/2022] Open
Abstract
Three-dimensional (3D) printing is a game changer technology that holds great promise for a wide variety of biomedical applications, including ophthalmology. Through this emerging technique, specific eye tissues can be custom-fabricated in a flexible and automated way, incorporating different cell types and biomaterials in precise anatomical 3D geometries. However, and despite the great progress and possibilities generated in recent years, there are still challenges to overcome that jeopardize its clinical application in regular practice. The main goal of this review is to provide an in-depth understanding of the current status and implementation of 3D bioprinting technology in the ophthalmology field in order to manufacture relevant tissues such as cornea, retina and conjunctiva. Special attention is paid to the description of the most commonly employed bioprinting methods, and the most relevant eye tissue engineering studies performed by 3D bioprinting technology at preclinical level. In addition, other relevant issues related to use of 3D bioprinting for ocular drug delivery, as well as both ethical and regulatory aspects, are analyzed. Through this review, we aim to raise awareness among the research community and report recent advances and future directions in order to apply this advanced therapy in the eye tissue regeneration field.
Collapse
Affiliation(s)
- Sandra Ruiz-Alonso
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (S.R.-A.); (I.V.-B.); (I.G.); (M.L.-M.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, 01009 Vitoria-Gasteiz, Spain
| | - Ilia Villate-Beitia
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (S.R.-A.); (I.V.-B.); (I.G.); (M.L.-M.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, 01009 Vitoria-Gasteiz, Spain
| | - Idoia Gallego
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (S.R.-A.); (I.V.-B.); (I.G.); (M.L.-M.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, 01009 Vitoria-Gasteiz, Spain
| | - Markel Lafuente-Merchan
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (S.R.-A.); (I.V.-B.); (I.G.); (M.L.-M.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, 01009 Vitoria-Gasteiz, Spain
| | - Gustavo Puras
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (S.R.-A.); (I.V.-B.); (I.G.); (M.L.-M.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, 01009 Vitoria-Gasteiz, Spain
| | - Laura Saenz-del-Burgo
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (S.R.-A.); (I.V.-B.); (I.G.); (M.L.-M.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, 01009 Vitoria-Gasteiz, Spain
- Correspondence: (L.S.-d.-B.); (J.L.P.); Tel.: +(34)-945014542 (L.S.-d.-B.); +(34)-945013091 (J.L.P.)
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (S.R.-A.); (I.V.-B.); (I.G.); (M.L.-M.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, 01009 Vitoria-Gasteiz, Spain
- Correspondence: (L.S.-d.-B.); (J.L.P.); Tel.: +(34)-945014542 (L.S.-d.-B.); +(34)-945013091 (J.L.P.)
| |
Collapse
|
29
|
Grolman JM, Singh M, Mooney DJ, Eriksson E, Nuutila K. Antibiotic-Containing Agarose Hydrogel for Wound and Burn Care. J Burn Care Res 2020; 40:900-906. [PMID: 31250003 DOI: 10.1093/jbcr/irz113] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Wound infections cause inflammation, tissue damage, and delayed healing that can lead to invasive infection and even death. The efficacy of systemic antibiotics is limited due to poor tissue penetration that is especially a problem in burn and blast wounds where the microcirculation is disrupted. Topical administration of antimicrobials is an attractive approach because it prevents infection and avoids systemic toxicity, while hydrogels are an appealing vehicle for topical drug delivery. They are easy to apply to the wound site by being injectable, the drug release properties can be controlled, and their many characteristics, such as biodegradation, mechanical strength, and chemical and biological response to stimuli can be tailored. Hydrogels also create a moist wound environment that is beneficial for healing. The purpose of this study was to formulate an agarose hydrogel that contains high concentrations of minocycline or gentamicin and study its characteristics. Subsequently, the minocycline agarose hydrogel was tested in a porcine burn model and its effect as a prophylactic treatment was studied. The results demonstrated that 0.5% agarose in water was the optimal concentration in terms of viscosity and pH. Bench testing at room temperature demonstrated that both antibiotics remained stable in the hydrogel for at least 7 days and both antibiotics demonstrated sustained release over the time of the experiment. The porcine burn experiment showed that prophylactic treatment with the agarose minocycline hydrogel decreased the burn depth and reduced the number of bacteria as efficiently as the commonly used silver sulfadiazine cream.
Collapse
Affiliation(s)
- Joshua M Grolman
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts.,John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Mansher Singh
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - David J Mooney
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts.,John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | | | - Kristo Nuutila
- Division of Plastic Surgery, Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
30
|
De Pieri A, Byerley AM, Musumeci CR, Salemizadehparizi F, Vanderhorst MA, Wuertz‐Kozak K. Electrospinning and 3D bioprinting for intervertebral disc tissue engineering. JOR Spine 2020; 3:e1117. [PMID: 33392454 PMCID: PMC7770193 DOI: 10.1002/jsp2.1117] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is a major cause of low back pain and represents a massive socioeconomic burden. Current conservative and surgical treatments fail to restore native tissue architecture and functionality. Tissue engineering strategies, especially those based on 3D bioprinting and electrospinning, have emerged as possible alternatives by producing cell-seeded scaffolds that replicate the structure of the IVD extracellular matrix. In this review, we provide an overview of recent advancements and limitations of 3D bioprinting and electrospinning for the treatment of IVD degeneration, focusing on future areas of research that may contribute to their clinical translation.
Collapse
Affiliation(s)
- Andrea De Pieri
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
| | - Ann M. Byerley
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
| | - Catherine R. Musumeci
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
| | | | - Maya A. Vanderhorst
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
| | - Karin Wuertz‐Kozak
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
- Schön Clinic Munich Harlaching, Spine CenterAcademic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (AU)MunichGermany
| |
Collapse
|
31
|
Ioannidis K, Danalatos RI, Champeris Tsaniras S, Kaplani K, Lokka G, Kanellou A, Papachristou DJ, Bokias G, Lygerou Z, Taraviras S. A Custom Ultra-Low-Cost 3D Bioprinter Supports Cell Growth and Differentiation. Front Bioeng Biotechnol 2020; 8:580889. [PMID: 33251196 PMCID: PMC7676439 DOI: 10.3389/fbioe.2020.580889] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/13/2020] [Indexed: 12/18/2022] Open
Abstract
Advances in 3D bioprinting have allowed the use of stem cells along with biomaterials and growth factors toward novel tissue engineering approaches. However, the cost of these systems along with their consumables is currently extremely high, limiting their applicability. To address this, we converted a 3D printer into an open source 3D bioprinter and produced a customized bioink based on accessible alginate/gelatin precursors, leading to a cost-effective solution. The bioprinter's resolution, including line width, spreading ratio and extrusion uniformity measurements, along with the rheological properties of the bioinks were analyzed, revealing high bioprinting accuracy within the printability window. Following the bioprinting process, cell survival and proliferation were validated on HeLa Kyoto and HEK293T cell lines. In addition, we isolated and 3D bioprinted postnatal neural stem cell progenitors derived from the mouse subventricular zone as well as mesenchymal stem cells derived from mouse bone marrow. Our results suggest that our low-cost 3D bioprinter can support cell proliferation and differentiation of two different types of primary stem cell populations, indicating that it can be used as a reliable tool for developing efficient research models for stem cell research and tissue engineering.
Collapse
Affiliation(s)
| | | | | | - Konstantina Kaplani
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| | - Georgia Lokka
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| | - Anastasia Kanellou
- Laboratory of Bone and Soft Tissue Studies, Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, Patras, Greece
| | - Dionysios J. Papachristou
- Laboratory of Bone and Soft Tissue Studies, Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, Patras, Greece
| | - Georgios Bokias
- Department of Chemistry, University of Patras, Patras, Greece
| | - Zoi Lygerou
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| |
Collapse
|
32
|
Sanz-Garcia A, Sodupe-Ortega E, Pernía-Espinoza A, Shimizu T, Escobedo-Lucea C. A Versatile Open-Source Printhead for Low-Cost 3D Microextrusion-Based Bioprinting. Polymers (Basel) 2020; 12:E2346. [PMID: 33066265 PMCID: PMC7602012 DOI: 10.3390/polym12102346] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional (3D) bioprinting promises to be essential in tissue engineering for solving the rising demand for organs and tissues. Some bioprinters are commercially available, but their impact on the field of Tissue engineering (TE) is still limited due to their cost or difficulty to tune. Herein, we present a low-cost easy-to-build printhead for microextrusion-based bioprinting (MEBB) that can be installed in many desktop 3D printers to transform them into 3D bioprinters. We can extrude bioinks with precise control of print temperature between 2-60 °C. We validated the versatility of the printhead, by assembling it in three low-cost open-source desktop 3D printers. Multiple units of the printhead can also be easily put together in a single printer carriage for building a multi-material 3D bioprinter. Print resolution was evaluated by creating representative calibration models at different temperatures using natural hydrogels such as gelatin and alginate, and synthetic ones like poloxamer. Using one of the three modified low-cost 3D printers, we successfully printed cell-laden lattice constructs with cell viabilities higher than 90% after 24-h post printing. Controlling temperature and pressure according to the rheological properties of the bioinks was essential in achieving optimal printability and great cell viability. The cost per unit of our device, which can be used with syringes of different volume, is less expensive than any other commercially available product. These data demonstrate an affordable open-source printhead with the potential to become a reliable alternative to commercial bioprinters for any laboratory.
Collapse
Affiliation(s)
- Andres Sanz-Garcia
- Division of Pharmaceutical Biosciences, University of Helsinki, Viikinkaari 5 E (P.O. Box 56), 00014 Helsinki, Finland; (A.S.-G.); (E.S.-O.)
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan;
| | - Enrique Sodupe-Ortega
- Division of Pharmaceutical Biosciences, University of Helsinki, Viikinkaari 5 E (P.O. Box 56), 00014 Helsinki, Finland; (A.S.-G.); (E.S.-O.)
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan;
- Department of Mechanical Engineering, University of La Rioja, San José de Calasanz 31, Edificio Departamental, 26004 Logroño, Spain;
| | - Alpha Pernía-Espinoza
- Department of Mechanical Engineering, University of La Rioja, San José de Calasanz 31, Edificio Departamental, 26004 Logroño, Spain;
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan;
| | - Carmen Escobedo-Lucea
- Division of Pharmaceutical Biosciences, University of Helsinki, Viikinkaari 5 E (P.O. Box 56), 00014 Helsinki, Finland; (A.S.-G.); (E.S.-O.)
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan;
| |
Collapse
|
33
|
Roche CD, Brereton RJL, Ashton AW, Jackson C, Gentile C. Current challenges in three-dimensional bioprinting heart tissues for cardiac surgery. Eur J Cardiothorac Surg 2020; 58:500-510. [PMID: 32391914 PMCID: PMC8456486 DOI: 10.1093/ejcts/ezaa093] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/27/2020] [Accepted: 02/18/2020] [Indexed: 12/25/2022] Open
Abstract
SUMMARY Previous attempts in cardiac bioengineering have failed to provide tissues for cardiac regeneration. Recent advances in 3-dimensional bioprinting technology using prevascularized myocardial microtissues as 'bioink' have provided a promising way forward. This review guides the reader to understand why myocardial tissue engineering is difficult to achieve and how revascularization and contractile function could be restored in 3-dimensional bioprinted heart tissue using patient-derived stem cells.
Collapse
Affiliation(s)
- Christopher D Roche
- Northern Clinical School of Medicine, University of Sydney, Kolling Institute, St Leonards, Sydney, NSW, Australia
- Department of Cardiothoracic Surgery, Royal North Shore Hospital, St Leonards, Sydney, NSW, Australia
- Department of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney (UTS), Ultimo, Sydney, NSW, Australia
- Department of Cardiothoracic Surgery, University Hospital of Wales, Cardiff, UK
| | - Russell J L Brereton
- Department of Cardiothoracic Surgery, Royal North Shore Hospital, St Leonards, Sydney, NSW, Australia
| | - Anthony W Ashton
- Northern Clinical School of Medicine, University of Sydney, Kolling Institute, St Leonards, Sydney, NSW, Australia
| | - Christopher Jackson
- Northern Clinical School of Medicine, University of Sydney, Kolling Institute, St Leonards, Sydney, NSW, Australia
| | - Carmine Gentile
- Northern Clinical School of Medicine, University of Sydney, Kolling Institute, St Leonards, Sydney, NSW, Australia
- Department of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney (UTS), Ultimo, Sydney, NSW, Australia
| |
Collapse
|
34
|
Florian DC, Odziomek M, Ock CL, Chen H, Guelcher SA. Principles of computer-controlled linear motion applied to an open-source affordable liquid handler for automated micropipetting. Sci Rep 2020; 10:13663. [PMID: 32788742 PMCID: PMC7424513 DOI: 10.1038/s41598-020-70465-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/13/2020] [Indexed: 11/09/2022] Open
Abstract
OTTO is an open-source automated liquid handler that can be fabricated at a cost of $1,500 using off-the-shelf and 3D-printable parts as an alternative to commercial devices. Open-source approaches have been applied to build syringe pumps, centrifuges, and other laboratory equipment. These devices are affordable but generally rely on a single motor to perform simple operations and thus do not fully utilize the potential of the Maker Movement. Open-source linear actuators and microcontrollers enable the fabrication of more complex laboratory instruments that rely on 3D positioning and accurate dispensing of fluids, such as automated liquid handlers. These instruments can be built rapidly and affordably, thereby providing access to highly reproducible sample preparation for common biological assays such as qPCR. We applied the design principles of speed and accuracy, unattended automation, and open-source components to build an automated liquid handler that controls micropipetting of liquids in 3D space at speeds and positional resolutions required for qPCR. In benchmarking studies, OTTO showed accuracy and sample preparation times comparable to manual qPCR. The ability to control linear motion and liquid dispensing using affordable off-the-shelf and 3D-printable parts can facilitate the adoption of open-source automated liquid handlers for qPCR, bioplotting, and other bioinstrumentation applications.
Collapse
Affiliation(s)
- David C Florian
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.,Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Mateusz Odziomek
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Cerie L Ock
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Hannah Chen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Scott A Guelcher
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA. .,Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
35
|
Soman SS, Vijayavenkataraman S. Applications of 3D Bioprinted-Induced Pluripotent Stem Cells in Healthcare. Int J Bioprint 2020; 6:280. [PMID: 33088994 PMCID: PMC7557348 DOI: 10.18063/ijb.v6i4.280] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology and advancements in three-dimensional (3D) bioprinting technology enable scientists to reprogram somatic cells to iPSCs and 3D print iPSC-derived organ constructs with native tissue architecture and function. iPSCs and iPSC-derived cells suspended in hydrogels (bioinks) allow to print tissues and organs for downstream medical applications. The bioprinted human tissues and organs are extremely valuable in regenerative medicine as bioprinting of autologous iPSC-derived organs eliminates the risk of immune rejection with organ transplants. Disease modeling and drug screening in bioprinted human tissues will give more precise information on disease mechanisms, drug efficacy, and drug toxicity than experimenting on animal models. Bioprinted iPSC-derived cancer tissues will aid in the study of early cancer development and precision oncology to discover patient-specific drugs. In this review, we present a brief summary of the combined use of two powerful technologies, iPSC technology, and 3D bioprinting in health-care applications.
Collapse
Affiliation(s)
- Soja Saghar Soman
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Sanjairaj Vijayavenkataraman
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE.,Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, NY, USA
| |
Collapse
|
36
|
Bioink Temperature Influence on Shear Stress, Pressure and Velocity Using Computational Simulation. Processes (Basel) 2020. [DOI: 10.3390/pr8070865] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Bioinks are usually cell-laden hydrogels widely studied in bioprinting performing experimental tests to tune their rheological properties, thus increasing research time and development costs. Computational Fluids Dynamics (CFD) is a powerful tool that can minimize iterations and costs simulating the material behavior using parametric changes in rheological properties under testing. Additionally, most bioinks have specific functionalities and their properties might widely change with temperature. Therefore, commercial bioinks are an excellent way to standardize bioprinting process, but they are not analyzed in detail. Therefore, the objective of this work is to study how three temperatures of the Cellink Bioink influence shear stress pressure and velocity through computational simulation. A comparison of three conical nozzles (20, 22, and 25G) for each temperature has been performed. The results show that shear stress, pressure, and velocity vary in negligible ranges for all combinations. Although these ranges are small and define a good thermo-responsive bioink, they do not generate a filament on the air and make drops during extrusion. In conclusion, this bioink provides a very stable behavior with low shear stress, but other bioprinting parameters must be set up to get a stable filament width.
Collapse
|
37
|
Extrusion-Based Bioprinting: Current Standards and Relevancy for Human-Sized Tissue Fabrication. Methods Mol Biol 2020; 2140:65-92. [PMID: 32207106 DOI: 10.1007/978-1-0716-0520-2_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The field of bioengineering has long pursued the goal of fabricating large-scale tissue constructs for use both in vitro and in vivo. Recent technological advances have indicated that bioprinting will be a key technique in manufacturing these specimens. This chapter aims to provide an overview of what has been achieved to date through the use of microextrusion bioprinters and what major challenges still impede progress. Microextrusion printer configurations will be addressed along with critical design characteristics including nozzle specifications and bioink modifications. Significant challenges within the field with regard to achieving long-term cell viability and vascularization, and current research that shows promise in mitigating these challenges in the near future are discussed. While microextrusion is a broad field with many applications, this chapter aims to provide an overview of the field with a focus on its applications toward human-sized tissue constructs.
Collapse
|
38
|
Salaris F, Colosi C, Brighi C, Soloperto A, de Turris V, Benedetti MC, Ghirga S, Rosito M, Di Angelantonio S, Rosa A. 3D Bioprinted Human Cortical Neural Constructs Derived from Induced Pluripotent Stem Cells. J Clin Med 2019; 8:E1595. [PMID: 31581732 PMCID: PMC6832547 DOI: 10.3390/jcm8101595] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/17/2023] Open
Abstract
Bioprinting techniques use bioinks made of biocompatible non-living materials and cells to build 3D constructs in a controlled manner and with micrometric resolution. 3D bioprinted structures representative of several human tissues have been recently produced using cells derived by differentiation of induced pluripotent stem cells (iPSCs). Human iPSCs can be differentiated in a wide range of neurons and glia, providing an ideal tool for modeling the human nervous system. Here we report a neural construct generated by 3D bioprinting of cortical neurons and glial precursors derived from human iPSCs. We show that the extrusion-based printing process does not impair cell viability in the short and long term. Bioprinted cells can be further differentiated within the construct and properly express neuronal and astrocytic markers. Functional analysis of 3D bioprinted cells highlights an early stage of maturation and the establishment of early network activity behaviors. This work lays the basis for generating more complex and faithful 3D models of the human nervous systems by bioprinting neural cells derived from iPSCs.
Collapse
Affiliation(s)
- Federico Salaris
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (F.S.); (C.C.); (C.B.); (A.S.); (V.d.T.); (S.G.); (M.R.)
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy;
| | - Cristina Colosi
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (F.S.); (C.C.); (C.B.); (A.S.); (V.d.T.); (S.G.); (M.R.)
| | - Carlo Brighi
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (F.S.); (C.C.); (C.B.); (A.S.); (V.d.T.); (S.G.); (M.R.)
| | - Alessandro Soloperto
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (F.S.); (C.C.); (C.B.); (A.S.); (V.d.T.); (S.G.); (M.R.)
| | - Valeria de Turris
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (F.S.); (C.C.); (C.B.); (A.S.); (V.d.T.); (S.G.); (M.R.)
| | - Maria Cristina Benedetti
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy;
| | - Silvia Ghirga
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (F.S.); (C.C.); (C.B.); (A.S.); (V.d.T.); (S.G.); (M.R.)
| | - Maria Rosito
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (F.S.); (C.C.); (C.B.); (A.S.); (V.d.T.); (S.G.); (M.R.)
| | - Silvia Di Angelantonio
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (F.S.); (C.C.); (C.B.); (A.S.); (V.d.T.); (S.G.); (M.R.)
- Department of Physiology and Pharmacology, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Alessandro Rosa
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (F.S.); (C.C.); (C.B.); (A.S.); (V.d.T.); (S.G.); (M.R.)
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy;
| |
Collapse
|
39
|
Mollica PA, Booth-Creech EN, Reid JA, Zamponi M, Sullivan SM, Palmer XL, Sachs PC, Bruno RD. 3D bioprinted mammary organoids and tumoroids in human mammary derived ECM hydrogels. Acta Biomater 2019; 95:201-213. [PMID: 31233891 DOI: 10.1016/j.actbio.2019.06.017] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 05/21/2019] [Accepted: 06/13/2019] [Indexed: 12/27/2022]
Abstract
The extracellular matrix (ECM) of tissues is an important mediator of cell function. Moreover, understanding cellular dynamics within their specific tissue context is also important for developmental biology, cancer research, and regenerative medicine. However, robust in vitro models that incorporate tissue-specific microenvironments are lacking. Here we describe a novel mammary-specific culture protocol that combines a self-gelling hydrogel comprised solely of ECM from decellularized rat or human breast tissue with the use of our previously described 3D bioprinting platform. We initially demonstrate that undigested and decellularized mammary tissue can support mammary epithelial and tumor cell growth. We then describe a methodology for generating mammary ECM extracts that can spontaneously gel to form hydrogels. These ECM hydrogels retain unique structural and signaling profiles that elicit differential responses when normal mammary and breast cancer cells are cultured within them. Using our bioprinter, we establish that we can generate large organoids/tumoroids in the all mammary-derived hydrogel. These findings demonstrate that our system allows for growth of organoids/tumoroids in a tissue-specific matrix with unique properties, thus providing a suitable platform for ECM and epithelial/cancer cell studies. STATEMENT OF SIGNIFICANCE: Factors within extracellular matrices (ECMs) are specific to their tissue of origin. It has been shown that tissue specific factors within the mammary gland's ECM have pronounced effects on cellular differentiation and cancer behavior. Understanding the role of the ECM in controlling cell fate has major implications for developmental biology, tissue engineering, and cancer therapy. However, in vitro models to study cellular interactions with tissue specific ECM are lacking. Here we describe the generation of 3D hydrogels consisting solely of human or mouse mammary ECM. We demonstrate that these novel 3D culture substrates can sustain large 3D bioprinted organoid and tumoroid formation. This is the first demonstration of an all mammary ECM culture system capable of sustaining large structural growths.
Collapse
|
40
|
Salaris F, Rosa A. Construction of 3D in vitro models by bioprinting human pluripotent stem cells: Challenges and opportunities. Brain Res 2019; 1723:146393. [PMID: 31425681 DOI: 10.1016/j.brainres.2019.146393] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/11/2019] [Accepted: 08/14/2019] [Indexed: 12/26/2022]
Abstract
Three-dimensional (3D) printing of biological material, or 3D bioprinting, is a rapidly expanding field with interesting applications in tissue engineering and regenerative medicine. Bioprinters use cells and biocompatible materials as an ink (bioink) to build 3D structures representative of organs and tissues, in a controlled manner and with micrometric resolution. Human embryonic (hESCs) and induced (hiPSCs) pluripotent stem cells are ideally able to provide all cell types found in the human body. A limited, but growing, number of recent reports suggest that cells derived by differentiation of hESCs and hiPSCs can be used as building blocks in bioprinted human 3D models, reproducing the cellular variety and cytoarchitecture of real tissues. In this review we will illustrate these examples, which include hepatic, cardiac, vascular, corneal and cartilage tissues, and discuss challenges and opportunities of bioprinting more demanding cell types, such as neurons, obtained from human pluripotent stem cells.
Collapse
Affiliation(s)
- Federico Salaris
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Alessandro Rosa
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy.
| |
Collapse
|
41
|
Romanazzo S, Nemec S, Roohani I. iPSC Bioprinting: Where are We at? MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2453. [PMID: 31374871 PMCID: PMC6696162 DOI: 10.3390/ma12152453] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 07/27/2019] [Accepted: 07/30/2019] [Indexed: 12/29/2022]
Abstract
Here, we present a concise review of current 3D bioprinting technologies applied to induced pluripotent stem cells (iPSC). iPSC have recently received a great deal of attention from the scientific and clinical communities for their unique properties, which include abundant adult cell sources, ability to indefinitely self-renew and differentiate into any tissue of the body. Bioprinting of iPSC and iPSC derived cells combined with natural or synthetic biomaterials to fabricate tissue mimicked constructs, has emerged as a technology that might revolutionize regenerative medicine and patient-specific treatment. This review covers the advantages and disadvantages of bioprinting techniques, influence of bioprinting parameters and printing condition on cell viability, and commonly used iPSC sources, and bioinks. A clear distinction is made for bioprinting techniques used for iPSC at their undifferentiated stage or when used as adult stem cells or terminally differentiated cells. This review presents state of the art data obtained from major searching engines, including Pubmed/MEDLINE, Google Scholar, and Scopus, concerning iPSC generation, undifferentiated iPSC, iPSC bioprinting, bioprinting techniques, cartilage, bone, heart, neural tissue, skin, and hepatic tissue cells derived from iPSC.
Collapse
Affiliation(s)
- Sara Romanazzo
- Biomaterials Design and Tissue Engineering Lab, School of Chemistry, University of New South Wales, New South Wales 2052, Australia
| | - Stephanie Nemec
- School of Materials Science and Engineering, University of New South Wales, New South Wales 2052, Australia
| | - Iman Roohani
- Biomaterials Design and Tissue Engineering Lab, School of Chemistry, University of New South Wales, New South Wales 2052, Australia.
| |
Collapse
|
42
|
Kahl M, Gertig M, Hoyer P, Friedrich O, Gilbert DF. Ultra-Low-Cost 3D Bioprinting: Modification and Application of an Off-the-Shelf Desktop 3D-Printer for Biofabrication. Front Bioeng Biotechnol 2019; 7:184. [PMID: 31417899 PMCID: PMC6684753 DOI: 10.3389/fbioe.2019.00184] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/15/2019] [Indexed: 01/01/2023] Open
Abstract
3D bioprinting has become a versatile and powerful method in tissue engineering and regenerative medicine and is increasingly adapted by other disciplines due to its tremendous potential beyond its typical applications. However, commercially available 3D bioprinting systems are typically expensive circumventing the broad implementation, including laboratories in low-resource settings. To address the limitations of conventional and commercially available technology, we developed a 3D bioprinter by modification of an off-the-shelf 3D desktop printer, that can be installed within a single day, is of handy size to fit into a standard laminar flow hood, customizable, ultra-low cost and thus, affordable to a broad range of research labs, or educational institutions. We evaluate accuracy and reproducibility of printing results using alginate and alginate/gelatin-hydrogels and demonstrate its potential for biomedical use by printing of various two-and three-dimensional cell-free and mammalian cell-laden objects using recombinant HEKYFP cells, stably expressing yellow fluorescent protein (YFP) as a model system and high-content imaging. We further provide a parts list and 3D design files in STL and STEP format for reconstructing the device. A time-lapse video of the custom-built device during operation is available at https://vimeo.com/274482794.
Collapse
Affiliation(s)
- Melanie Kahl
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Markus Gertig
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Phillipp Hoyer
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Daniel F Gilbert
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
43
|
Bruno RD, Reid J, Sachs PC. The revolution will be open-source: how 3D bioprinting can change 3D cell culture. Oncotarget 2019; 10:4724-4726. [PMID: 31523386 PMCID: PMC6730589 DOI: 10.18632/oncotarget.27099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 12/24/2022] Open
Affiliation(s)
- Robert D Bruno
- School of Medical Diagnostic & Translational Sciences, Old Dominion University, Norfolk, VA, USA
| | - John Reid
- School of Medical Diagnostic & Translational Sciences, Old Dominion University, Norfolk, VA, USA
| | - Patrick C Sachs
- School of Medical Diagnostic & Translational Sciences, Old Dominion University, Norfolk, VA, USA
| |
Collapse
|
44
|
A 3D bioprinter platform for mechanistic analysis of tumoroids and chimeric mammary organoids. Sci Rep 2019; 9:7466. [PMID: 31097753 PMCID: PMC6522494 DOI: 10.1038/s41598-019-43922-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 05/02/2019] [Indexed: 12/30/2022] Open
Abstract
The normal mammary microenvironment can suppress tumorigenesis and redirect cancer cells to adopt a normal mammary epithelial cell fate in vivo. Understanding of this phenomenon offers great promise for novel treatment and detection strategies in cancer, but current model systems make mechanistic insights into the process difficult. We have recently described a low-cost bioprinting platform designed to be accessible for basic cell biology laboratories. Here we report the use of this system for the study of tumorigenesis and microenvironmental redirection of breast cancer cells. We show our bioprinter significantly increases tumoroid formation in 3D collagen gels and allows for precise generation of tumoroid arrays. We also demonstrate that we can mimic published in vivo findings by co-printing cancer cells along with normal mammary epithelial cells to generate chimeric organoids. These chimeric organoids contain cancer cells that take part in normal luminal formation. Furthermore, we show for the first time that cancer cells within chimeric structures have a significant increase in 5-hydroxymethylcytosine levels as compared to bioprinted tumoroids. These results demonstrate the capacity of our 3D bioprinting platform to study tumorigenesis and microenvironmental control of breast cancer and highlight a novel mechanistic insight into the process of microenvironmental control of cancer.
Collapse
|
45
|
Evaluation of the Applicability of 3d Models as Perceived by the Students of Health Sciences. J Med Syst 2019; 43:108. [PMID: 30887131 DOI: 10.1007/s10916-019-1238-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
Abstract
The methodology and style of teaching anatomy in the faculties of Health Sciences is evolving due to the changes being introduced as a result of the application of new technologies. This brings a more positive attitude in the students, enabling an active participation during the lessons. One of these new technologies is the creation of 3D models that reliably recreates the anatomical details of real bone pieces and allow access of anatomy students to bone pieces that are not damaged and possess easily identifiable anatomical details. In our work, we have presented previously created 3D models of skull and jaw to the students of anatomy in the Faculties of Health Sciences of the University of Salamanca, Spain. The faculties included were odontology, medicine, occupational therapy nursing, health sciences and physiotherapy. A survey was carried out to assess the usefulness of these 3D models in the practical study of anatomy. The total number of students included in the survey was 280.The analysis of the results presents a positive evaluation about the use of 3D models by the students studying anatomy in different Faculties of Health Sciences.
Collapse
|
46
|
Okubo N, Qureshi A, Dalgarno K, Goh KL, Derebail S. Cost-effective microvalve-assisted bioprinter for tissue engineering. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.bprint.2019.e00043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
Reid JA, Mollica PA, Bruno RD, Sachs PC. Consistent and reproducible cultures of large-scale 3D mammary epithelial structures using an accessible bioprinting platform. Breast Cancer Res 2018; 20:122. [PMID: 30305139 PMCID: PMC6180647 DOI: 10.1186/s13058-018-1045-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 08/24/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Standard three-dimensional (3D) in vitro culture techniques, such as those used for mammary epithelial cells, rely on random distribution of cells within hydrogels. Although these systems offer advantages over traditional 2D models, limitations persist owing to the lack of control over cellular placement within the hydrogel. This results in experimental inconsistencies and random organoid morphology. Robust, high-throughput experimentation requires greater standardization of 3D epithelial culture techniques. METHODS Here, we detail the use of a 3D bioprinting platform as an investigative tool to control the 3D formation of organoids through the "self-assembly" of human mammary epithelial cells. Experimental bioprinting procedures were optimized to enable the formation of controlled arrays of individual mammary organoids. We define the distance and cell number parameters necessary to print individual organoids that do not interact between print locations as well as those required to generate large contiguous organoids connected through multiple print locations. RESULTS We demonstrate that as few as 10 cells can be used to form 3D mammary structures in a single print and that prints up to 500 μm apart can fuse to form single large structures. Using these fusion parameters, we demonstrate that both linear and non-linear (contiguous circles) can be generated with sizes of 3 mm in length/diameter. We confirm that cells from individual prints interact to form structures with a contiguous lumen. Finally, we demonstrate that organoids can be printed into human collagen hydrogels, allowing for all-human 3D culture systems. CONCLUSIONS Our platform is adaptable to different culturing protocols and is superior to traditional random 3D culture techniques in efficiency, reproducibility, and scalability. Importantly, owing to the low-cost accessibility and computer numerical control-driven platform of our 3D bioprinter, we have the ability to disseminate our experiments with absolute precision to interested laboratories.
Collapse
Affiliation(s)
- John A Reid
- Biomedical Engineering Institute, College of Engineering, Old Dominion University, 5115 Hampton Blvd, Norfolk, VA, 23529, USA
| | - Peter A Mollica
- School of Medical Diagnostic & Translational Sciences, College of Health Sciences, Old Dominion University, 5115 Hampton Blvd, Norfolk, VA, 23529, USA
| | - Robert D Bruno
- School of Medical Diagnostic & Translational Sciences, College of Health Sciences, Old Dominion University, 5115 Hampton Blvd, Norfolk, VA, 23529, USA.
| | - Patrick C Sachs
- School of Medical Diagnostic & Translational Sciences, College of Health Sciences, Old Dominion University, 5115 Hampton Blvd, Norfolk, VA, 23529, USA.
| |
Collapse
|
48
|
Ding S, Feng L, Wu J, Zhu F, Tan Z, Yao R. Bioprinting of Stem Cells: Interplay of Bioprinting Process, Bioinks, and Stem Cell Properties. ACS Biomater Sci Eng 2018; 4:3108-3124. [PMID: 33435052 DOI: 10.1021/acsbiomaterials.8b00399] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Combining the advantages of 3D bioprinting technology and biological characteristics of stem cells, bioprinting of stem cells is recognized as a novel technology with broad applications in biological study, drug testing, tissue engineering, regenerative medicine, etc. However, the biological performance and functional reconstruction of stem cells are greatly influenced by both the bioprinting process and post-bioprinting culture conditions, which are critical factors to consider for further applications. Here we review the recent development of stem cell bioprinting technology and conclude on the major factors regulating stem cell viability, proliferation, differentiation, and function from the aspects of the choice of bioprinting techniques, the modulation of bioprinting parameters, and the regulation of the stem cell niche in the whole lifespan of bioprinting practices. We aim to provide a comprehensive consideration and guidance regarding the bioprinting of stem cells for optimization of this promising technology in biological and medical applications.
Collapse
Affiliation(s)
- Supeng Ding
- Department of Mechanical Engineering, Biomanufacturing and Rapid Forming Technology, Key Laboratory of Beijing, Tsinghua University, Beijing 100084, People's Republic of China.,Department of Materials Science and Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Lu Feng
- Department of Mechanical Engineering, Biomanufacturing and Rapid Forming Technology, Key Laboratory of Beijing, Tsinghua University, Beijing 100084, People's Republic of China
| | - Jiayang Wu
- Department of Mechanical Engineering, Biomanufacturing and Rapid Forming Technology, Key Laboratory of Beijing, Tsinghua University, Beijing 100084, People's Republic of China.,Department of Construction Management, Tsinghua University, Beijing 100084, People's Republic of China
| | - Fei Zhu
- Department of Mechanical Engineering, Biomanufacturing and Rapid Forming Technology, Key Laboratory of Beijing, Tsinghua University, Beijing 100084, People's Republic of China
| | - Ze'en Tan
- Department of Mechanical Engineering, Biomanufacturing and Rapid Forming Technology, Key Laboratory of Beijing, Tsinghua University, Beijing 100084, People's Republic of China
| | - Rui Yao
- Department of Mechanical Engineering, Biomanufacturing and Rapid Forming Technology, Key Laboratory of Beijing, Tsinghua University, Beijing 100084, People's Republic of China
| |
Collapse
|
49
|
Ma X, Liu J, Zhu W, Tang M, Lawrence N, Yu C, Gou M, Chen S. 3D bioprinting of functional tissue models for personalized drug screening and in vitro disease modeling. Adv Drug Deliv Rev 2018; 132:235-251. [PMID: 29935988 PMCID: PMC6226327 DOI: 10.1016/j.addr.2018.06.011] [Citation(s) in RCA: 226] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 05/04/2018] [Accepted: 06/18/2018] [Indexed: 02/08/2023]
Abstract
3D bioprinting is emerging as a promising technology for fabricating complex tissue constructs with tailored biological components and mechanical properties. Recent advances have enabled scientists to precisely position materials and cells to build functional tissue models for in vitro drug screening and disease modeling. This review presents state-of-the-art 3D bioprinting techniques and discusses the choice of cell source and biomaterials for building functional tissue models that can be used for personalized drug screening and disease modeling. In particular, we focus on 3D-bioprinted liver models, cardiac tissues, vascularized constructs, and cancer models for their promising applications in medical research, drug discovery, toxicology, and other pre-clinical studies.
Collapse
Affiliation(s)
- Xuanyi Ma
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Justin Liu
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Wei Zhu
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Min Tang
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Natalie Lawrence
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Claire Yu
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, PR China
| | - Shaochen Chen
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, PR China.
| |
Collapse
|
50
|
Datta P, Barui A, Wu Y, Ozbolat V, Moncal KK, Ozbolat IT. Essential steps in bioprinting: From pre- to post-bioprinting. Biotechnol Adv 2018; 36:1481-1504. [PMID: 29909085 DOI: 10.1016/j.biotechadv.2018.06.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 05/15/2018] [Accepted: 06/10/2018] [Indexed: 12/17/2022]
Abstract
An increasing demand for directed assembly of biomaterials has inspired the development of bioprinting, which facilitates the assembling of both cellular and acellular inks into well-arranged three-dimensional (3D) structures for tissue fabrication. Although great advances have been achieved in the recent decade, there still exist issues to be addressed. Herein, a review has been systematically performed to discuss the considerations in the entire procedure of bioprinting. Though bioprinting is advancing at a rapid pace, it is seen that the whole process of obtaining tissue constructs from this technique involves multiple-stages, cutting across various technology domains. These stages can be divided into three broad categories: pre-bioprinting, bioprinting and post-bioprinting. Each stage can influence others and has a bearing on the performance of fabricated constructs. For example, in pre-bioprinting, tissue biopsy and cell expansion techniques are essential to ensure a large number of cells are available for mass organ production. Similarly, medical imaging is needed to provide high resolution designs, which can be faithfully bioprinted. In the bioprinting stage, compatibility of biomaterials is needed to be matched with solidification kinetics to ensure constructs with high cell viability and fidelity are obtained. On the other hand, there is a need to develop bioprinters, which have high degrees of freedom of movement, perform without failure concerns for several hours and are compact, and affordable. Finally, maturation of bioprinted cells are governed by conditions provided during the post-bioprinting process. This review, for the first time, puts all the bioprinting stages in perspective of the whole process of bioprinting, and analyzes their current state-of-the art. It is concluded that bioprinting community will recognize the relative importance and optimize the parameter of each stage to obtain the desired outcomes.
Collapse
Affiliation(s)
- Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah 711103, West Bengal, India
| | - Ananya Barui
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah 711103, West Bengal, India
| | - Yang Wu
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Veli Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; Ceyhan Engineering Faculty, Cukurova University, Adana 01950, Turkey
| | - Kazim K Moncal
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA; Materials Research Institute, Penn State University, University Park, PA 16802, USA.
| |
Collapse
|