1
|
Liu G, Xue J, Zhou X, Gui M, Xia R, Zhang Y, Cai Y, Li S, Shi S, Mao X, Chen Z. The paradigm shifts of periodontal regeneration strategy: From reparative manipulation to developmental engineering. Bioact Mater 2025; 49:418-436. [PMID: 40165829 PMCID: PMC11957753 DOI: 10.1016/j.bioactmat.2025.03.009] [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: 11/02/2024] [Revised: 02/07/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Ideal periodontal regeneration requires the integration of alveolar bone, periodontal ligament, and cementum, along with Sharpey's fibers for occlusal force resistance. However, physiological regeneration remains rare due to its intricate structure, making clinical regeneration a challenge. Periodontal ligament stem cells (PDLSCs), first isolated in 2004, hold the key to multi-directional differentiation into cementoblasts, fibroblasts, and osteoblasts. While traditional therapies like guided tissue regeneration (GTR) aim to activate PDLSCs, clinical outcomes are inconsistent, suggesting the need for additional strategies to enhance PDLSCs' functions. Advancements in molecular biotechnology have introduced the use of recombinant growth factors for tissue regeneration. However, maintaining their efficacy requires high doses, posing cost and safety issues. Multi-layered scaffolds combined with cell sheet technology offer new insights, but face production, ethical, and survival challenges. Immune regulation plays a crucial role in PDLSC-mediated regeneration. The concept of "coagulo-immunomodulation" has emerged, emphasizing the coupling of blood coagulation and immune responses for periodontal regeneration. Despite its potential, the clinical translation of immune-based strategies remains elusive. The "developmental engineering" approach, which mimics developmental events using embryonic-stage cells and microenvironments, shows promise. Our research group has made initial strides, indicating its potential as a viable solution for periodontal complex regeneration. However, further clinical trials and considerations are needed for successful clinical application. This review aims to summarize the strategic transitions in the development of periodontal regenerative materials and to propose prospective avenues for future development.
Collapse
Affiliation(s)
- Guanqi Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Junlong Xue
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Xuan Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Mixiao Gui
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Ruidi Xia
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Yanshu Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Yihua Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Shuhua Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Songtao Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- South China Center of Craniofacial Stem Cell Research, Guangzhou, 510055, China
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- South China Center of Craniofacial Stem Cell Research, Guangzhou, 510055, China
| | - Zetao Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| |
Collapse
|
2
|
Ikram M, Mahmud MAP, Kalyar AA, Alomayri T, Almahri A, Hussain D. 3D-bioprinting of MXenes: Developments, medical applications, challenges, and future roadmap. Colloids Surf B Biointerfaces 2025; 251:114568. [PMID: 40020571 DOI: 10.1016/j.colsurfb.2025.114568] [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: 11/18/2024] [Revised: 02/03/2025] [Accepted: 02/15/2025] [Indexed: 03/03/2025]
Abstract
MXenes is a member of 2D transition metals carbides and nitrides with promising application prospects in energy storage, sensing, nanomedicine, tissue engineering, catalysis, and electronics. In the current era, MXenes have been widely applied in biomedical applications due to their unique rheological and electrochemical attributes. They have a larger surface area with more active sites, higher conductivity, lower cytotoxicity, and greater biocompatibility, making them highly suitable candidates for in-vivo biomedical applications. Due to recent advancemnets in MXenes 3D bioprinting, they are widely applied in regenerative medicine to combat challenges in suitable transplantation of tissues and organs. However, 3D bioprinting of MXenes has several complexities based on cell type, cytotoxicity, cell viability, and differentiation. To address these intricacies, surface modifications of MXene materials are done, which makes them highly fascinating for the 3D printing of tissues and organs. In the current review, we summarized recent progress in 3D bioprinting of MXene materials to construct scaffolds with desired rheological and biological properties, focusing on their potential applications in cancer phototherapy, tissue engineering, bone regeneration, and biosensing. We also discussed parameters affecting their biomedical applications and possible solutions by applying surface modifications. In addition, we addressed current challenges and future roadmaps for 3D bioprinting of MXene materials, such as generating high throughput 3D printed tissue constructs, drug delivery, drug discovery, and toxicology.
Collapse
Affiliation(s)
- Muhammad Ikram
- Department of Agricultural and Biosystems Engineering, South Dakota State University, Brookings, SD 57007, United States of America.
| | - M A Parvez Mahmud
- School of Mathematical and Physical Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Amina Akbar Kalyar
- Department of Zoology, Wildlife and Fisheries, University of Agriculture Faisalabad, Pakistan
| | - Thamer Alomayri
- Department of Physics, Faculty of Science, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Albandary Almahri
- Department of Chemistry, College of Science and Humanities, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Dilshad Hussain
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
3
|
Lu C, Jin A, Liu H, Gao C, Sun W, Zhang Y, Dai Q, Liu Y. Advancing tissue engineering through vascularized cell spheroids: building blocks of the future. Biomater Sci 2025; 13:1901-1922. [PMID: 40067332 DOI: 10.1039/d4bm01206b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Vascularization is a crucial aspect of biofabrication, as the development of vascular networks is essential for tissue survival and the optimization of cellular functions. Spheroids have emerged as versatile units for vascularization, demonstrating significant potential in angiogenesis and prevascularization for tissue engineering and regenerative medicine. However, a major challenge in creating customized vascularized spheroids is the construction of a biomimetic extracellular matrix (ECM) microenvironment. This process requires careful regulation of environmental factors, including the modulation of growth factors, the selection of culture media, and the co-culture of diverse cell types. Recent advancements in biofabrication have expanded the potential applications of vascularized spheroids. The integration of microfluidic technology with bioprinting offers promising solutions to existing challenges in regenerative medicine. Spheroids have been widely studied for their ability to promote vascularization in in vitro models. This review highlights the latest developments in vascularized biofabrication, and systematically explores strategies for constructing vascularized spheroids. We provide a comprehensive analysis of spheroid applications in specific tissues, including skin, liver, bone, cardiac, and tumor models. Finally, the review addresses the major challenges and future directions in the field.
Collapse
Affiliation(s)
- Chunxiang Lu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Aoxiang Jin
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Huazhen Liu
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Chuang Gao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Wenbin Sun
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Yi Zhang
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Qiqi Dai
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai, 200444, China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
| |
Collapse
|
4
|
Tanvir MAH, Khaleque MA, Lee J, Park JB, Kim GH, Lee HH, Kim YY. Three-Dimensional Bioprinting for Intervertebral Disc Regeneration. J Funct Biomater 2025; 16:105. [PMID: 40137384 PMCID: PMC11943008 DOI: 10.3390/jfb16030105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
The rising demand for organ transplants and the need for precise tissue models have positioned the in vitro biomanufacturing of tissues and organs as a pivotal area in regenerative treatment. Considerable development has been achieved in growing tissue-engineered intervertebral disc (IVD) scaffolds, designed to meet stringent mechanical and biological compatibility criteria. Among the cutting-edge approaches, 3D bioprinting stands out due to its unparalleled capacity to organize biomaterials, bioactive molecules, and living cells with high precision. Despite these advancements, polymer-based scaffolds still encounter limitations in replicating the extracellular matrix (ECM)-like environment, which is fundamental for optimal cellular activities. To overcome these challenges, integrating polymers with hydrogels has been recommended as a promising solution. This combination enables the advancement of porous scaffolds that nurture cell adhesion, proliferation, as well as differentiation. Additionally, bioinks derived from the decellularized extracellular matrix (dECM) have exhibited potential in replicating biologically relevant microenvironments, enhancing cell viability, differentiation, and motility. Hydrogels, whether derived from natural sources involving collagen and alginate or synthesized chemically, are highly valued for their ECM-like properties and superior biocompatibility. This review will explore recent advancements in techniques and technologies for IVD regeneration. Emphasis will be placed on identifying research gaps and proposing strategies to bridge them, with the goal of accelerating the translation of IVDs into clinical applications.
Collapse
Affiliation(s)
- Md Amit Hasan Tanvir
- Department of Orthopedic Surgery, Daejeon St. Mary’s Hospital, The Catholic University of Korea, Seoul 34943, Republic of Korea; (M.A.H.T.); (M.A.K.); (G.-H.K.)
| | - Md Abdul Khaleque
- Department of Orthopedic Surgery, Daejeon St. Mary’s Hospital, The Catholic University of Korea, Seoul 34943, Republic of Korea; (M.A.H.T.); (M.A.K.); (G.-H.K.)
| | - Junhee Lee
- Department of Bionic Machinery, KIMM Institute of AI Robot, Korea Institute of Machinery and Materials, Daejeon 34103, Republic of Korea;
| | - Jong-Beom Park
- Department of Orthopedic Surgery, Uijeongbu Saint Mary’s Hospital, The Catholic University of Korea, Seoul 11765, Republic of Korea;
| | - Ga-Hyun Kim
- Department of Orthopedic Surgery, Daejeon St. Mary’s Hospital, The Catholic University of Korea, Seoul 34943, Republic of Korea; (M.A.H.T.); (M.A.K.); (G.-H.K.)
| | - Hwan-Hee Lee
- Department of Orthopedic Surgery, Daejeon St. Mary’s Hospital, The Catholic University of Korea, Seoul 34943, Republic of Korea; (M.A.H.T.); (M.A.K.); (G.-H.K.)
| | - Young-Yul Kim
- Department of Orthopedic Surgery, Daejeon St. Mary’s Hospital, The Catholic University of Korea, Seoul 34943, Republic of Korea; (M.A.H.T.); (M.A.K.); (G.-H.K.)
| |
Collapse
|
5
|
Xiang Y, Bao X, Sun T. Evaluation of bacteriophages for the alleviation of potential bacterial contamination risks in developmental engineering. Biotechnol Bioeng 2024; 121:3211-3223. [PMID: 39382053 DOI: 10.1002/bit.28778] [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: 02/21/2023] [Revised: 05/15/2024] [Accepted: 06/10/2024] [Indexed: 10/10/2024]
Abstract
This research aimed to address the potential bacterial contamination risks in developmental engineering (DE) using bacteriophages. To compare and contrast the exemplar Escherichia coli T4 and M13 bacteriophages, human dermal fibroblasts cultivated on culture plates, natural cellulosic scaffolds, and poly(methyl methacrylate) (PMMA) particles were utilized as two-dimensional (2D) cell, three-dimensional (3D) tissue, and modular tissue culture models, respectively. When directly introduced into these distinct culture systems, both phages survived, exhibited no significant effects on the cultured cells or tissues, yet displayed their potentials to alleviate the infections caused by corresponding bacterial host cells. Apart from direct addition into the culture medium, both phages were also coated on PMMA, polystyrene, poly(lactic acid) particles with different diameters (5, 10, 30, and 100 µm) and cellulosic scaffolds. The coated phages endured the coating processes and demonstrated their viabilities in plaque assays. Further testing indicated that the phages coated on the PMMA particles tolerated multiple deliberate rinses and centrifugations, but not thermal treatment at 60-80°C. In summary, T4 and M13 bacteriophages not only manifested their antibacterial functions in diverse 2D cell, 3D tissue, and modular tissue culture systems, but also demonstrated their potentials of coating modular scaffolds to alleviate the bacterial contamination risks in DE.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Materials, Loughborough University, Loughborough, UK
| | - Xujin Bao
- Department of Materials, Loughborough University, Loughborough, UK
| | - Tao Sun
- Department of Chemical Engineering, Loughborough University, Loughborough, UK
| |
Collapse
|
6
|
Gharibshahian M, Torkashvand M, Bavisi M, Aldaghi N, Alizadeh A. Recent advances in artificial intelligent strategies for tissue engineering and regenerative medicine. Skin Res Technol 2024; 30:e70016. [PMID: 39189880 PMCID: PMC11348508 DOI: 10.1111/srt.70016] [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: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Tissue engineering and regenerative medicine (TERM) aim to repair or replace damaged or lost tissues or organs due to accidents, diseases, or aging, by applying different sciences. For this purpose, an essential part of TERM is the designing, manufacturing, and evaluating of scaffolds, cells, tissues, and organs. Artificial intelligence (AI) or the intelligence of machines or software can be effective in all areas where computers play a role. METHODS The "artificial intelligence," "machine learning," "tissue engineering," "clinical evaluation," and "scaffold" keywords used for searching in various databases and articles published from 2000 to 2024 were evaluated. RESULTS The combination of tissue engineering and AI has created a new generation of technological advancement in the biomedical industry. Experience in TERM has been refined using advanced design and manufacturing techniques. Advances in AI, particularly deep learning, offer an opportunity to improve scientific understanding and clinical outcomes in TERM. CONCLUSION The findings of this research show the high potential of AI, machine learning, and robots in the selection, design, and fabrication of scaffolds, cells, tissues, or organs, and their analysis, characterization, and evaluation after their implantation. AI can be a tool to accelerate the introduction of tissue engineering products to the bedside. HIGHLIGHTS The capabilities of artificial intelligence (AI) can be used in different ways in all the different stages of TERM and not only solve the existing limitations, but also accelerate the processes, increase efficiency and precision, reduce costs, and complications after transplantation. ML predicts which technologies have the most efficient and easiest path to enter the market and clinic. The use of AI along with these imaging techniques can lead to the improvement of diagnostic information, the reduction of operator errors when reading images, and the improvement of image analysis (such as classification, localization, regression, and segmentation).
Collapse
Affiliation(s)
- Maliheh Gharibshahian
- Nervous System Stem Cells Research CenterSemnan University of Medical SciencesSemnanIran
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineSemnan University of Medical SciencesSemnanIran
| | | | - Mahya Bavisi
- Department of Tissue Engineering and Applied Cell SciencesSchool of Advanced Technologies in MedicineIran University of Medical SciencesTehranIran
| | - Niloofar Aldaghi
- Student Research CommitteeSchool of MedicineShahroud University of Medical SciencesShahroudIran
| | - Akram Alizadeh
- Nervous System Stem Cells Research CenterSemnan University of Medical SciencesSemnanIran
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineSemnan University of Medical SciencesSemnanIran
| |
Collapse
|
7
|
Baecher H, Hoch CC, Knoedler S, Maheta BJ, Kauke-Navarro M, Safi AF, Alfertshofer M, Knoedler L. From bench to bedside - current clinical and translational challenges in fibula free flap reconstruction. Front Med (Lausanne) 2023; 10:1246690. [PMID: 37886365 PMCID: PMC10598714 DOI: 10.3389/fmed.2023.1246690] [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: 06/24/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023] Open
Abstract
Fibula free flaps (FFF) represent a working horse for different reconstructive scenarios in facial surgery. While FFF were initially established for mandible reconstruction, advancements in planning for microsurgical techniques have paved the way toward a broader spectrum of indications, including maxillary defects. Essential factors to improve patient outcomes following FFF include minimal donor site morbidity, adequate bone length, and dual blood supply. Yet, persisting clinical and translational challenges hamper the effectiveness of FFF. In the preoperative phase, virtual surgical planning and artificial intelligence tools carry untapped potential, while the intraoperative role of individualized surgical templates and bioprinted prostheses remains to be summarized. Further, the integration of novel flap monitoring technologies into postoperative patient management has been subject to translational and clinical research efforts. Overall, there is a paucity of studies condensing the body of knowledge on emerging technologies and techniques in FFF surgery. Herein, we aim to review current challenges and solution possibilities in FFF. This line of research may serve as a pocket guide on cutting-edge developments and facilitate future targeted research in FFF.
Collapse
Affiliation(s)
- Helena Baecher
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Cosima C. Hoch
- Medical Faculty, Friedrich Schiller University Jena, Jena, Germany
| | - Samuel Knoedler
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bhagvat J. Maheta
- College of Medicine, California Northstate University, Elk Grove, CA, United States
| | - Martin Kauke-Navarro
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Ali-Farid Safi
- Craniologicum, Center for Cranio-Maxillo-Facial Surgery, Bern, Switzerland
- Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Michael Alfertshofer
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
8
|
Tripathi S, Mandal SS, Bauri S, Maiti P. 3D bioprinting and its innovative approach for biomedical applications. MedComm (Beijing) 2023; 4:e194. [PMID: 36582305 PMCID: PMC9790048 DOI: 10.1002/mco2.194] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 12/26/2022] Open
Abstract
3D bioprinting or additive manufacturing is an emerging innovative technology revolutionizing the field of biomedical applications by combining engineering, manufacturing, art, education, and medicine. This process involved incorporating the cells with biocompatible materials to design the required tissue or organ model in situ for various in vivo applications. Conventional 3D printing is involved in constructing the model without incorporating any living components, thereby limiting its use in several recent biological applications. However, this uses additional biological complexities, including material choice, cell types, and their growth and differentiation factors. This state-of-the-art technology consciously summarizes different methods used in bioprinting and their importance and setbacks. It also elaborates on the concept of bioinks and their utility. Biomedical applications such as cancer therapy, tissue engineering, bone regeneration, and wound healing involving 3D printing have gained much attention in recent years. This article aims to provide a comprehensive review of all the aspects associated with 3D bioprinting, from material selection, technology, and fabrication to applications in the biomedical fields. Attempts have been made to highlight each element in detail, along with the associated available reports from recent literature. This review focuses on providing a single platform for cancer and tissue engineering applications associated with 3D bioprinting in the biomedical field.
Collapse
Affiliation(s)
- Swikriti Tripathi
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Subham Shekhar Mandal
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Sudepta Bauri
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Pralay Maiti
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| |
Collapse
|
9
|
Introduction to three-dimensional printing in medicine. 3D Print Med 2023. [DOI: 10.1016/b978-0-323-89831-7.00008-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
10
|
Guo JL, Januszyk M, Longaker MT. Machine Learning in Tissue Engineering. Tissue Eng Part A 2023; 29:2-19. [PMID: 35943870 PMCID: PMC9885550 DOI: 10.1089/ten.tea.2022.0128] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/02/2022] [Indexed: 02/03/2023] Open
Abstract
Machine learning (ML) and artificial intelligence have accelerated scientific discovery, augmented clinical practice, and deepened fundamental understanding of many biological phenomena. ML technologies have now been applied to diverse areas of tissue engineering research, including biomaterial design, scaffold fabrication, and cell/tissue modeling. Emerging ML-empowered strategies include machine-optimized polymer synthesis, predictive modeling of scaffold fabrication processes, complex analyses of structure-function relationships, and deep learning of spatialized cell phenotypes and tissue composition. The emergence of ML in tissue engineering, while relatively recent, has already enabled increasingly complex and multivariate analyses of the relationships between biological, chemical, and physical factors in driving tissue regenerative outcomes. This review highlights the novel methodologies, emerging strategies, and areas of potential growth within this rapidly evolving area of research. Impact statement Machine learning (ML) has accelerated scientific discovery and augmented clinical practice across multiple fields. Now, ML has driven exciting new paradigms in tissue engineering research, including machine-optimized biomaterial design, predictive modeling of scaffold fabrication, and spatiotemporal analysis of cell and tissue systems. The emergence of ML in tissue engineering, while relatively recent, has already enabled increasingly complex analyses of the relationships between biological, chemical, and physical factors in driving tissue regenerative outcomes. This review highlights the novel methodologies, emerging strategies, and areas of potential growth within this rapidly evolving area of research.
Collapse
Affiliation(s)
- Jason L. Guo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael Januszyk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
11
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|
12
|
Arjoca S, Robu A, Neagu M, Neagu A. Mathematical and computational models in spheroid-based biofabrication. Acta Biomater 2022:S1742-7061(22)00418-4. [PMID: 35853599 DOI: 10.1016/j.actbio.2022.07.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/25/2022] [Accepted: 07/12/2022] [Indexed: 11/01/2022]
Abstract
Ubiquitous in embryonic development, tissue fusion is of interest to tissue engineers who use tissue spheroids or organoids as building blocks of three-dimensional (3D) multicellular constructs. This review presents mathematical models and computer simulations of the fusion of tissue spheroids. The motivation of this study stems from the need to predict the post-printing evolution of 3D bioprinted constructs. First, we provide a brief overview of differential adhesion, the main morphogenetic mechanism involved in post-printing structure formation. It will be shown that clusters of cohesive cells behave as an incompressible viscous fluid on the time scale of hours. The discussion turns then to mathematical models based on the continuum hydrodynamics of highly viscous liquids and on statistical mechanics. Next, we analyze the validity and practical use of computational models of multicellular self-assembly in live constructs created by tissue spheroid bioprinting. Finally, we discuss the perspectives of the field as machine learning starts to reshape experimental design, and modular robotic workstations tend to alleviate the burden of repetitive tasks in biofabrication. STATEMENT OF SIGNIFICANCE: Bioprinted constructs are living systems, which evolve via morphogenetic mechanisms known from developmental biology. This review presents mathematical and computational tools devised for modeling post-printing structure formation. They help achieving a desirable outcome without expensive optimization experiments. While previous reviews mainly focused on assumptions, technical details, strengths, and limitations of computational models of multicellular self-assembly, this article discusses their validity and practical use in biofabrication. It also presents an overview of mathematical models that proved to be useful in the evaluation of experimental data on tissue spheroid fusion, and in the calibration of computational models. Finally, the perspectives of the field are discussed in the advent of robotic biofabrication platforms and bioprinting process optimization by machine learning.
Collapse
Affiliation(s)
- Stelian Arjoca
- Center for Modeling Biological Systems and Data Analysis, Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, Piata Eftimie Murgu Nr. 2-4, Timisoara 300041, Romania
| | - Andreea Robu
- Department of Automation and Applied Informatics, Politehnica University of Timisoara, Timisoara 300006, Romania
| | - Monica Neagu
- Center for Modeling Biological Systems and Data Analysis, Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, Piata Eftimie Murgu Nr. 2-4, Timisoara 300041, Romania
| | - Adrian Neagu
- Center for Modeling Biological Systems and Data Analysis, Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, Piata Eftimie Murgu Nr. 2-4, Timisoara 300041, Romania; Department of Physics & Astronomy, University of Missouri-Columbia, Columbia, MO 65211, USA.
| |
Collapse
|
13
|
Chen Z, Zhou B, Wang X, Zhou G, Zhang W, Yi B, Wang W, Liu W. Synergistic effects of mechanical stimulation and crimped topography to stimulate natural collagen development for tendon engineering. Acta Biomater 2022; 145:297-315. [PMID: 35470072 DOI: 10.1016/j.actbio.2022.04.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/06/2022] [Accepted: 04/14/2022] [Indexed: 12/28/2022]
Abstract
Suitable scaffold structures and mechanical loading are essential for functional tendon engineering. However, the bipolar fibril structure of native tendon collagen is yet to be recaptured in engineered tendons. This study compared the development of Achilles tendons of postnatal rats with and without (via surgical section) mechanical loading to define the mechanism of mechanical stimulation-mediated tendon development. The results demonstrated that the severed tendons weakened mechanically and exhibited disorganization without a bipolar fibril superstructure. Proteomic analysis revealed differentially expressed key regulatory molecules related to the collagen assembly process, including decreased fibromodulin, keratocan, fibroblast growth factor-1, and increased lumican and collagen5a1 in the severed tendons with immunohistochemical verification. Additionally, a complex regulatory network of mechanical stimulation-mediated collagen assembly in a spatiotemporal manner was also revealed using bioinformatics analysis, wherein PI3K-Akt and HDAC4 may be the predominant signaling pathways. A wavy microgrooved surface (Y = 5.47sin(0.015x)) that biomimics tendon topography was observed to enhance the expression of collagen assembly molecules under mechanical loading, and the aforementioned pathways are particularly involved and verified with their respective inhibitors of LY-294002 and LMK-235. Furthermore, an electrospun crimped nanofiber scaffold (approximately 2 μm fiber diameter and 0.12 crimpness) was fabricated to biomimic the tenogenic niche environment; this was observed to be more effective on enhancing collagen production and assembly under mechanical stimulation. In conclusion, the synergistic effect between topographical niche and mechanical stimulation was observed to be essential for collagen assembly and maturation and should be applied to functional tendon engineering in the future. STATEMENT OF SIGNIFICANCE: In biomaterial-mediated tendon regeneration, mechanical stimulation is essential for tendon collagen assembly. However, the underlying mechanisms remain not fully defined, leading to the failure of the native-like collagen regeneration. In this study, a mechanical stimulation deprivation model of rat tendon was established to reveal the mechanisms in tendon development and define the key regulatory molecules including small leucine-rich proteoglycans, lysyl oxidase and collagen V. After ensuring the importance of biomimetic structure in tendon remodeling, crimped nanofibers were developed to verify these regulatory molecules, and demonstrated that mechanical stimulation significantly enhanced collagen assembly via PIK3 and HDAC4 pathways in biomaterial-regulated tendon regeneration. This study provides more insightful perspectives in the physiologically remodeling progression of tendon collagen and design of tendon scaffolds.
Collapse
Affiliation(s)
- Zhenying Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Tissue Engineering Research, Shanghai 200011, China
| | - Boya Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Tissue Engineering Research, Shanghai 200011, China
| | - Xiansong Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Tissue Engineering Research, Shanghai 200011, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Tissue Engineering Research, Shanghai 200011, China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Tissue Engineering Research, Shanghai 200011, China
| | - Bingcheng Yi
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Tissue Engineering Research, Shanghai 200011, China.
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Tissue Engineering Research, Shanghai 200011, China.
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Tissue Engineering Research, Shanghai 200011, China.
| |
Collapse
|
14
|
Kasamkattil J, Gryadunova A, Martin I, Barbero A, Schären S, Krupkova O, Mehrkens A. Spheroid-Based Tissue Engineering Strategies for Regeneration of the Intervertebral Disc. Int J Mol Sci 2022; 23:2530. [PMID: 35269672 PMCID: PMC8910276 DOI: 10.3390/ijms23052530] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Degenerative disc disease, a painful pathology of the intervertebral disc (IVD), often causes disability and reduces quality of life. Although regenerative cell-based strategies have shown promise in clinical trials, none have been widely adopted clinically. Recent developments demonstrated that spheroid-based approaches might help overcome challenges associated with cell-based IVD therapies. Spheroids are three-dimensional multicellular aggregates with architecture that enables the cells to differentiate and synthesize endogenous ECM, promotes cell-ECM interactions, enhances adhesion, and protects cells from harsh conditions. Spheroids could be applied in the IVD both in scaffold-free and scaffold-based configurations, possibly providing advantages over cell suspensions. This review highlights areas of future research in spheroid-based regeneration of nucleus pulposus (NP) and annulus fibrosus (AF). We also discuss cell sources and methods for spheroid fabrication and characterization, mechanisms related to spheroid fusion, as well as enhancement of spheroid performance in the context of the IVD microenvironment.
Collapse
Affiliation(s)
- Jesil Kasamkattil
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; (J.K.); (A.G.); (S.S.); (A.M.)
| | - Anna Gryadunova
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; (J.K.); (A.G.); (S.S.); (A.M.)
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (I.M.); (A.B.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (I.M.); (A.B.)
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (I.M.); (A.B.)
| | - Stefan Schären
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; (J.K.); (A.G.); (S.S.); (A.M.)
| | - Olga Krupkova
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; (J.K.); (A.G.); (S.S.); (A.M.)
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (I.M.); (A.B.)
- Lepage Research Institute, University of Prešov, 17. Novembra 1, 081 16 Prešov, Slovakia
| | - Arne Mehrkens
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; (J.K.); (A.G.); (S.S.); (A.M.)
| |
Collapse
|
15
|
Ramadan Q, Zourob M. 3D Bioprinting at the Frontier of Regenerative Medicine, Pharmaceutical, and Food Industries. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 2:607648. [PMID: 35047890 PMCID: PMC8757855 DOI: 10.3389/fmedt.2020.607648] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/08/2020] [Indexed: 12/22/2022] Open
Abstract
3D printing technology has emerged as a key driver behind an ongoing paradigm shift in the production process of various industrial domains. The integration of 3D printing into tissue engineering, by utilizing life cells which are encapsulated in specific natural or synthetic biomaterials (e.g., hydrogels) as bioinks, is paving the way toward devising many innovating solutions for key biomedical and healthcare challenges and heralds' new frontiers in medicine, pharmaceutical, and food industries. Here, we present a synthesis of the available 3D bioprinting technology from what is found and what has been achieved in various applications and discussed the capabilities and limitations encountered in this technology.
Collapse
Affiliation(s)
- Qasem Ramadan
- College of Science and General Studies, Alfaisal University, Riyadh, Saudi Arabia
| | - Mohammed Zourob
- College of Science and General Studies, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Bertassoni LE. Bioprinting of Complex Multicellular Organs with Advanced Functionality-Recent Progress and Challenges Ahead. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2101321. [PMID: 35060652 PMCID: PMC10171718 DOI: 10.1002/adma.202101321] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/20/2021] [Indexed: 05/12/2023]
Abstract
Bioprinting has emerged as one of the most promising strategies for fabrication of functional organs in the lab as an alternative to transplant organs. While progress in the field has mostly been restricted to a few miniaturized tissues with minimal biological functionality until a few years ago, recent progress has advanced the concept of building three-dimensional multicellular organ complexity remarkably. This review discusses a series of milestones that have paved the way for bioprinting of tissue constructs that have advanced levels of biological and architectural functionality. Critical materials, engineering and biological challenges that are key to addressing the desirable function of engineered organs are presented. These are discussed in light of the many difficulties to replicate the heterotypic organization of multicellular solid organs, the nanoscale precision of the extracellular microenvironment in hierarchical tissues, as well as the advantages and limitations of existing bioprinting methods to adequately overcome these barriers. In summary, the advances of the field toward realistic manufacturing of functional organs have never been so extensive, and this manuscript serves as a road map for some of the recent progress and the challenges ahead.
Collapse
Affiliation(s)
- Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, School of Dentistry, Oregon Health and Science University, Portland, OR, 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
- Center for Regenerative Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
- Cancer Early Detection Advanced Research (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97239, USA
| |
Collapse
|
17
|
A Paradigm Shift in Tissue Engineering: From a Top–Down to a Bottom–Up Strategy. Processes (Basel) 2021. [DOI: 10.3390/pr9060935] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Tissue engineering (TE) was initially designed to tackle clinical organ shortage problems. Although some engineered tissues have been successfully used for non-clinical applications, very few (e.g., reconstructed human skin) have been used for clinical purposes. As the current TE approach has not achieved much success regarding more broad and general clinical applications, organ shortage still remains a challenging issue. This very limited clinical application of TE can be attributed to the constraints in manufacturing fully functional tissues via the traditional top–down approach, where very limited cell types are seeded and cultured in scaffolds with equivalent sizes and morphologies as the target tissues. The newly proposed developmental engineering (DE) strategy towards the manufacture of fully functional tissues utilises a bottom–up approach to mimic developmental biology processes by implementing gradual tissue assembly alongside the growth of multiple cell types in modular scaffolds. This approach may overcome the constraints of the traditional top–down strategy as it can imitate in vivo-like tissue development processes. However, several essential issues must be considered, and more mechanistic insights of the fundamental, underpinning biological processes, such as cell–cell and cell–material interactions, are necessary. The aim of this review is to firstly introduce and compare the number of cell types, the size and morphology of the scaffolds, and the generic tissue reconstruction procedures utilised in the top–down and the bottom–up strategies; then, it will analyse their advantages, disadvantages, and challenges; and finally, it will briefly discuss the possible technologies that may overcome some of the inherent limitations of the bottom–up strategy.
Collapse
|
18
|
Orsolits B, Kovács Z, Kriston-Vizi J, Merkely B, Földes G. New Modalities of 3D Pluripotent Stem Cell-Based Assays in Cardiovascular Toxicity. Front Pharmacol 2021; 12:603016. [PMID: 33854431 PMCID: PMC8039822 DOI: 10.3389/fphar.2021.603016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/04/2021] [Indexed: 12/04/2022] Open
Abstract
The substantial progress of the human induced pluripotent stem cell (hiPSC) technologies over the last decade has provided us with new opportunities for cardiovascular drug discovery, regenerative medicine, and disease modeling. The combination of hiPSC with 3D culture techniques offers numerous advantages for generating and studying physiological and pathophysiological cardiac models. Cells grown in 3D can overcome many limitations of 2D cell cultures and animal models. Furthermore, it enables the investigation in an architecturally appropriate, complex cellular environment in vitro. Yet, generation and study of cardiac organoids-which may contain versatile cardiovascular cell types differentiated from hiPSC-remain a challenge. The large-scale and high-throughput applications require accurate and standardised models with highly automated processes in culturing, imaging and data collection. Besides the compound spatial structure of organoids, their biological processes also possess different temporal dynamics which require other methods and technologies to detect them. In this review, we summarise the possibilities and challenges of acquiring relevant information from 3D cardiovascular models. We focus on the opportunities during different time-scale processes in dynamic pharmacological experiments and discuss the putative steps toward one-size-fits-all assays.
Collapse
Affiliation(s)
- Barbara Orsolits
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
| | - Zsófia Kovács
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
| | - János Kriston-Vizi
- Bioinformatics Image Core (BIONIC), MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
| | - Gábor Földes
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
- National Heart and Lung Institute, Imperial Centre for Experimental and Translational Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
19
|
Contessi Negrini N, Angelova Volponi A, Higgins C, Sharpe P, Celiz A. Scaffold-based developmental tissue engineering strategies for ectodermal organ regeneration. Mater Today Bio 2021; 10:100107. [PMID: 33889838 PMCID: PMC8050778 DOI: 10.1016/j.mtbio.2021.100107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/15/2021] [Accepted: 02/27/2021] [Indexed: 12/12/2022] Open
Abstract
Tissue engineering (TE) is a multidisciplinary research field aiming at the regeneration, restoration, or replacement of damaged tissues and organs. Classical TE approaches combine scaffolds, cells and soluble factors to fabricate constructs mimicking the native tissue to be regenerated. However, to date, limited success in clinical translations has been achieved by classical TE approaches, because of the lack of satisfactory biomorphological and biofunctional features of the obtained constructs. Developmental TE has emerged as a novel TE paradigm to obtain tissues and organs with correct biomorphology and biofunctionality by mimicking the morphogenetic processes leading to the tissue/organ generation in the embryo. Ectodermal appendages, for instance, develop in vivo by sequential interactions between epithelium and mesenchyme, in a process known as secondary induction. A fine artificial replication of these complex interactions can potentially lead to the fabrication of the tissues/organs to be regenerated. Successful developmental TE applications have been reported, in vitro and in vivo, for ectodermal appendages such as teeth, hair follicles and glands. Developmental TE strategies require an accurate selection of cell sources, scaffolds and cell culture configurations to allow for the correct replication of the in vivo morphogenetic cues. Herein, we describe and discuss the emergence of this TE paradigm by reviewing the achievements obtained so far in developmental TE 3D scaffolds for teeth, hair follicles, and salivary and lacrimal glands, with particular focus on the selection of biomaterials and cell culture configurations.
Collapse
Affiliation(s)
| | - A. Angelova Volponi
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - C.A. Higgins
- Department of Bioengineering, Imperial College London, London, UK
| | - P.T. Sharpe
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - A.D. Celiz
- Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
20
|
Zohorsky K, Mequanint K. Designing Biomaterials to Modulate Notch Signaling in Tissue Engineering and Regenerative Medicine. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:383-410. [PMID: 33040694 DOI: 10.1089/ten.teb.2020.0182] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The design of cell-instructive biomaterials for tissue engineering and regenerative medicine is at a crossroads. Although the conventional tissue engineering approach is top-down (cells seeded to macroporous scaffolds and mature to form tissues), bottom-up tissue engineering strategies are becoming appealing. With such developments, we can study cell signaling events, thus enabling functional tissue assembly in physiologic and diseased models. Among many important signaling pathways, the Notch signaling pathway is the most diverse in its influence during tissue morphogenesis and repair following injury. Although Notch signaling is extensively studied in developmental biology and cancer biology, our knowledge of designing biomaterial-based Notch signaling platforms and incorporating Notch signaling components into engineered tissue systems is limited. By incorporating Notch signaling to tissue engineering scaffolds, we can direct cell-specific responses and improve engineered tissue maturation. This review will discuss recent progress in the development of Notch signaling biomaterials as a promising target to control cellular fate decisions, including the influences of ligand identity, biophysical material cues, ligand presentation strategies, and mechanotransduction. Notch signaling is consequently of interest to direct, control, and reprogram cellular behavior on a biomaterial surface. We anticipate that discussions in this article will allow for enhanced knowledge and insight into designing Notch targeted biomaterials for various tissue engineering and cell fate determinations. Impact statement Notch signaling is recognized as an important pathway in tissue engineering and regenerative medicine; however, there is no systematic review on this topic. The comprehensive review and perspectives presented here provide an in-depth discussion on ligand presentation strategies both in 2D and in 3D cell culture environments involving biomaterials/scaffolds. In addition, this review article provides insight into the challenges in designing cell surrogate biomaterials capable of providing Notch signals. To the best of the authors' knowledge, this is the first review relevant to the fields of tissue engineering.
Collapse
Affiliation(s)
- Kathleen Zohorsky
- School of Biomedical Engineering and The University of Western Ontario, London, Canada
| | - Kibret Mequanint
- School of Biomedical Engineering and The University of Western Ontario, London, Canada.,Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Canada
| |
Collapse
|
21
|
Kosheleva NV, Efremov YM, Shavkuta BS, Zurina IM, Zhang D, Zhang Y, Minaev NV, Gorkun AA, Wei S, Shpichka AI, Saburina IN, Timashev PS. Cell spheroid fusion: beyond liquid drops model. Sci Rep 2020; 10:12614. [PMID: 32724115 PMCID: PMC7387529 DOI: 10.1038/s41598-020-69540-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/19/2020] [Indexed: 01/14/2023] Open
Abstract
Biological self-assembly is crucial in the processes of development, tissue regeneration, and maturation of bioprinted tissue-engineered constructions. The cell aggregates-spheroids-have become widely used model objects in the study of this phenomenon. Existing approaches describe the fusion of cell aggregates by analogy with the coalescence of liquid droplets and ignore the complex structural properties of spheroids. Here, we analyzed the fusion process in connection with structure and mechanical properties of the spheroids from human somatic cells of different phenotypes: mesenchymal stem cells from the limbal eye stroma and epithelial cells from retinal pigment epithelium. A nanoindentation protocol was applied for the mechanical measurements. We found a discrepancy with the liquid drop fusion model: the fusion was faster for spheroids from epithelial cells with lower apparent surface tension than for mesenchymal spheroids with higher surface tension. This discrepancy might be caused by biophysical processes such as extracellular matrix remodeling in the case of mesenchymal spheroids and different modes of cell migration. The obtained results will contribute to the development of more realistic models for spheroid fusion that would further provide a helpful tool for constructing cell aggregates with required properties both for fundamental studies and tissue reparation.
Collapse
Affiliation(s)
- Nastasia V Kosheleva
- FSBSI "Institute of General Pathology and Pathophysiology", 8, Baltiyskaya st., Moscow, 125315, Russia.
- FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of Russia, 2/1, Barrikadnaya St., Moscow, 125993, Russia.
- Faculty of Biology, Lomonosov Moscow State University, 12-1, Leninskie Gory, Moscow, 119234, Russia.
| | - Yuri M Efremov
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow, 119991, Russia
| | - Boris S Shavkuta
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow, 119991, Russia
- Institute of Photonic Technologies, Research Center "Crystallography and Photonics" RAS, 2, Pionerskaya st., Troitsk, Moscow, 142190, Russia
| | - Irina M Zurina
- FSBSI "Institute of General Pathology and Pathophysiology", 8, Baltiyskaya st., Moscow, 125315, Russia
- FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of Russia, 2/1, Barrikadnaya St., Moscow, 125993, Russia
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow, 119991, Russia
| | - Deying Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yuanyuan Zhang
- Wake Forest University Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Nikita V Minaev
- Institute of Photonic Technologies, Research Center "Crystallography and Photonics" RAS, 2, Pionerskaya st., Troitsk, Moscow, 142190, Russia
| | - Anastasiya A Gorkun
- FSBSI "Institute of General Pathology and Pathophysiology", 8, Baltiyskaya st., Moscow, 125315, Russia
- FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of Russia, 2/1, Barrikadnaya St., Moscow, 125993, Russia
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow, 119991, Russia
| | - Shicheng Wei
- Department of Oral and Maxillofacial Surgery/Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Anastasia I Shpichka
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow, 119991, Russia
| | - Irina N Saburina
- FSBSI "Institute of General Pathology and Pathophysiology", 8, Baltiyskaya st., Moscow, 125315, Russia
- FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of Russia, 2/1, Barrikadnaya St., Moscow, 125993, Russia
| | - Peter S Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow, 119991, Russia
- Institute of Photonic Technologies, Research Center "Crystallography and Photonics" RAS, 2, Pionerskaya st., Troitsk, Moscow, 142190, Russia
- Department of Polymers and Composites, N.N. Semenov Institute of Chemical Physics, 4, Kosygin st., Moscow, 119991, Russia
- Chemistry Department, Lomonosov Moscow State University, 1‑3, Leninskiye Gory, Moscow, 119991, Russia
| |
Collapse
|
22
|
Effects of Cryopreservation on Cell Metabolic Activity and Function of Biofabricated Structures Laden with Osteoblasts. MATERIALS 2020; 13:ma13081966. [PMID: 32331435 PMCID: PMC7215951 DOI: 10.3390/ma13081966] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
Abstract
Biofabrication and maturation of bone constructs is a long-term task that requires a high degree of specialization. This specialization falls onto the hierarchy complexity of the bone tissue that limits the transfer of this technology to the clinic. This work studied the effects of the short-term cryopreservation on biofabricated osteoblast-containing structures, with the final aim to make them steadily available in biobanks. The biological responses studied include the osteoblast post-thawing metabolic activity and the recovery of the osteoblastic function of 3D-bioprinted osteoblastic structures and beta tricalcium phosphate (β-TCP) scaffolds infiltrated with osteoblasts encapsulated in a hydrogel. The obtained structures were cryopreserved at −80 °C for 7 days using dimethyl sulfoxide (DMSO) as cryoprotectant additive. After thawing the structures were cultured up to 14 days. The results revealed fundamental biological aspects for the successful cryopreservation of osteoblast constructs. In summary, immature osteoblasts take longer to recover than mature osteoblasts. The pre-cryopreservation culture period had an important effect on the metabolic activity and function maintain, faster recovering normal values when cryopreserved after longer-term culture (7 days). The use of β-TCP scaffolds further improved the osteoblast survival after cryopreservation, resulting in similar levels of alkaline phosphatase activity in comparison with the non-preserved structures. These results contribute to the understanding of the biology of cryopreserved osteoblast constructs, approaching biofabrication to the clinical practice.
Collapse
|
23
|
Zubillaga V, Alonso-Varona A, Fernandes SCM, Salaberria AM, Palomares T. Adipose-Derived Mesenchymal Stem Cell Chondrospheroids Cultured in Hypoxia and a 3D Porous Chitosan/Chitin Nanocrystal Scaffold as a Platform for Cartilage Tissue Engineering. Int J Mol Sci 2020; 21:E1004. [PMID: 32028724 PMCID: PMC7037297 DOI: 10.3390/ijms21031004] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 01/06/2023] Open
Abstract
Articular cartilage degeneration is one of the most common causes of pain and disability in middle-aged and older people. Tissue engineering (TE) has shown great therapeutic promise for this condition. The design of cartilage regeneration constructs must take into account the specific characteristics of the cartilaginous matrix, as well as the avascular nature of cartilage and its cells' peculiar arrangement in isogenic groups. Keeping these factors in mind, we have designed a 3D porous scaffold based on genipin-crosslinked chitosan/chitin nanocrystals for spheroid chondral differentiation of human adipose tissue-derived mesenchymal stem cells (hASCs) induced in hypoxic conditions. First, we demonstrated that, under low oxygen conditions, the chondrospheroids obtained express cartilage-specific markers including collagen type II (COL2A1) and aggrecan, lacking expression of osteogenic differentiation marker collagen type I (COL1A2). These results were associated with an increased expression of hypoxia-inducible factor 1α, which positively directs COL2A1 and aggrecan expression. Finally, we determined the most suitable chondrogenic differentiation pattern when hASC spheroids were seeded in the 3D porous scaffold under hypoxia and obtained a chondral extracellular matrix with a high sulphated glycosaminoglycan content, which is characteristic of articular cartilage. These findings highlight the potential use of such templates in cartilage tissue engineering.
Collapse
Affiliation(s)
- Veronica Zubillaga
- Department of Cell Biology and Histology, Faculty of Medicine and Nursey, University of the Basque Country (UPV/EHU), B Sarriena s/n, 48940 Leioa, Spain;
| | - Ana Alonso-Varona
- Department of Cell Biology and Histology, Faculty of Medicine and Nursey, University of the Basque Country (UPV/EHU), B Sarriena s/n, 48940 Leioa, Spain;
| | - Susana C. M. Fernandes
- Institute of Analytical Sciences and Physico-chemistry for the Environment and Materials, University of Pau and Pays Adour, E2S UPPA, CNRS, 64600 Anglet, France;
| | - Asier M. Salaberria
- Biorefinery Processes Research Group, Department of Chemical and Environmental Engineering, Polytechnic School, University of the Basque Country (UPV/EHU), Pza. Europa 1, 20018 Donostia-San Sebastian, Spain;
| | - Teodoro Palomares
- Department of Surgery, Radiology and Physic Medicine, Faculty of Medicine, University of the Basque Country (UPV/EHU), B Sarriena, s/n, 48940 Leioa, Spain
| |
Collapse
|
24
|
Ma Z, Holle AW, Melde K, Qiu T, Poeppel K, Kadiri VM, Fischer P. Acoustic Holographic Cell Patterning in a Biocompatible Hydrogel. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904181. [PMID: 31782570 DOI: 10.1002/adma.201904181] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/11/2019] [Indexed: 05/21/2023]
Abstract
Acoustophoresis is promising as a rapid, biocompatible, noncontact cell manipulation method, where cells are arranged along the nodes or antinodes of the acoustic field. Typically, the acoustic field is formed in a resonator, which results in highly symmetric regular patterns. However, arbitrary, nonsymmetrically shaped cell assemblies are necessary to obtain the irregular cellular arrangements found in biological tissues. It is shown that arbitrarily shaped cell patterns can be obtained from the complex acoustic field distribution defined by an acoustic hologram. Attenuation of the sound field induces localized acoustic streaming and the resultant convection flow gently delivers the suspended cells to the image plane where they form the designed pattern. It is shown that the process can be implemented in a biocompatible collagen solution, which can then undergo gelation to immobilize the cell pattern inside the viscoelastic matrix. The patterned cells exhibit F-actin-based protrusions, which indicate that the cells grow and thrive within the matrix. Cell viability assays and brightfield imaging after one week confirm cell survival and that the patterns persist. Acoustophoretic cell manipulation by holographic fields thus holds promise for noncontact, long-range, long-term cellular pattern formation, with a wide variety of potential applications in tissue engineering and mechanobiology.
Collapse
Affiliation(s)
- Zhichao Ma
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569, Stuttgart, Germany
| | - Andrew W Holle
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg, 69120, Germany
| | - Kai Melde
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569, Stuttgart, Germany
| | - Tian Qiu
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569, Stuttgart, Germany
- Institute of Physical Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569, Stuttgart, Germany
| | - Korbinian Poeppel
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569, Stuttgart, Germany
| | - Vincent Mauricio Kadiri
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569, Stuttgart, Germany
- Institute of Physical Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569, Stuttgart, Germany
| | - Peer Fischer
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569, Stuttgart, Germany
- Institute of Physical Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569, Stuttgart, Germany
| |
Collapse
|
25
|
Rana Khalid I, Darakhshanda I, Rafi a R. 3D Bioprinting: An attractive alternative to traditional organ transplantation. ACTA ACUST UNITED AC 2019. [DOI: 10.17352/abse.000012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
26
|
Andrique L, Recher G, Alessandri K, Pujol N, Feyeux M, Bon P, Cognet L, Nassoy P, Bikfalvi A. A model of guided cell self-organization for rapid and spontaneous formation of functional vessels. SCIENCE ADVANCES 2019; 5:eaau6562. [PMID: 31206014 PMCID: PMC6561743 DOI: 10.1126/sciadv.aau6562] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 05/10/2019] [Indexed: 05/15/2023]
Abstract
Most achievements to engineer blood vessels are based on multiple-step manipulations such as manual sheet rolling or sequential cell seeding followed by scaffold degradation. Here, we propose a one-step strategy using a microfluidic coextrusion device to produce mature functional blood vessels. A hollow alginate hydrogel tube is internally coated with extracellular matrix to direct the self-assembly of a mixture of endothelial cells (ECs) and smooth muscle cells (SMCs). The resulting vascular structure has the correct configuration of lumen, an inner lining of ECs, and outer sheath of SMCs. These "vesseloids" reach homeostasis within a day and exhibit the following properties expected for functional vessels (i) quiescence, (ii) perfusability, and (iii) contractility in response to vasoconstrictor agents. Together, these findings provide an original and simple strategy to generate functional artificial vessels and pave the way for further developments in vascular graft and tissue engineering and for deciphering the angiogenesis process.
Collapse
Affiliation(s)
- L. Andrique
- LAMC, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers (Inserm U1029) F-33170 Pessac, France
- Université de Bordeaux, F-33170 Pessac, France
| | - G. Recher
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400 Talence, France
- Institut d’Optique Graduate School & CNRS UMR 5298, F-33400 Talence, France
| | - K. Alessandri
- Institut d’Optique Graduate School & CNRS UMR 5298, F-33400 Talence, France
| | - N. Pujol
- LAMC, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers (Inserm U1029) F-33170 Pessac, France
- Université de Bordeaux, F-33170 Pessac, France
| | - M. Feyeux
- Institut d’Optique Graduate School & CNRS UMR 5298, F-33400 Talence, France
| | - P. Bon
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400 Talence, France
- Institut d’Optique Graduate School & CNRS UMR 5298, F-33400 Talence, France
| | - L. Cognet
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400 Talence, France
- Institut d’Optique Graduate School & CNRS UMR 5298, F-33400 Talence, France
| | - P. Nassoy
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400 Talence, France
- Institut d’Optique Graduate School & CNRS UMR 5298, F-33400 Talence, France
| | - A. Bikfalvi
- LAMC, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers (Inserm U1029) F-33170 Pessac, France
- Université de Bordeaux, F-33170 Pessac, France
| |
Collapse
|
27
|
Wragg NM, Burke L, Wilson SL. A critical review of current progress in 3D kidney biomanufacturing: advances, challenges, and recommendations. RENAL REPLACEMENT THERAPY 2019. [DOI: 10.1186/s41100-019-0218-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
28
|
Sarker M, Naghieh S, McInnes AD, Schreyer DJ, Chen X. Regeneration of peripheral nerves by nerve guidance conduits: Influence of design, biopolymers, cells, growth factors, and physical stimuli. Prog Neurobiol 2018; 171:125-150. [DOI: 10.1016/j.pneurobio.2018.07.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 01/10/2023]
|
29
|
Sarker M, Naghieh S, Sharma N, Chen X. 3D biofabrication of vascular networks for tissue regeneration: A report on recent advances. J Pharm Anal 2018; 8:277-296. [PMID: 30345141 PMCID: PMC6190507 DOI: 10.1016/j.jpha.2018.08.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/24/2018] [Accepted: 08/26/2018] [Indexed: 12/19/2022] Open
Abstract
Rapid progress in tissue engineering research in past decades has opened up vast possibilities to tackle the challenges of generating tissues or organs that mimic native structures. The success of tissue engineered constructs largely depends on the incorporation of a stable vascular network that eventually anastomoses with the host vasculature to support the various biological functions of embedded cells. In recent years, significant progress has been achieved with respect to extrusion, laser, micro-molding, and electrospinning-based techniques that allow the fabrication of any geometry in a layer-by-layer fashion. Moreover, decellularized matrix, self-assembled structures, and cell sheets have been explored to replace the biopolymers needed for scaffold fabrication. While the techniques have evolved to create specific tissues or organs with outstanding geometric precision, formation of interconnected, functional, and perfused vascular networks remains a challenge. This article briefly reviews recent progress in 3D fabrication approaches used to fabricate vascular networks with incorporated cells, angiogenic factors, proteins, and/or peptides. The influence of the fabricated network on blood vessel formation, and the various features, merits, and shortcomings of the various fabrication techniques are discussed and summarized.
Collapse
Affiliation(s)
- M.D. Sarker
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Saman Naghieh
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - N.K. Sharma
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
30
|
Lepowsky E, Muradoglu M, Tasoglu S. Towards preserving post-printing cell viability and improving the resolution: Past, present, and future of 3D bioprinting theory. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.bprint.2018.e00034] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
31
|
Tasnim N, De la Vega L, Anil Kumar S, Abelseth L, Alonzo M, Amereh M, Joddar B, Willerth SM. 3D Bioprinting Stem Cell Derived Tissues. Cell Mol Bioeng 2018; 11:219-240. [PMID: 31719887 PMCID: PMC6816617 DOI: 10.1007/s12195-018-0530-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Stem cells offer tremendous promise for regenerative medicine as they can become a variety of cell types. They also continuously proliferate, providing a renewable source of cells. Recently, it has been found that 3D printing constructs using stem cells, can generate models representing healthy or diseased tissues, as well as substitutes for diseased and damaged tissues. Here, we review the current state of the field of 3D printing stem cell derived tissues. First, we cover 3D printing technologies and discuss the different types of stem cells used for tissue engineering applications. We then detail the properties required for the bioinks used when printing viable tissues from stem cells. We give relevant examples of such bioprinted tissues, including adipose tissue, blood vessels, bone, cardiac tissue, cartilage, heart valves, liver, muscle, neural tissue, and pancreas. Finally, we provide future directions for improving the current technologies, along with areas of focus for future work to translate these exciting technologies into clinical applications.
Collapse
Affiliation(s)
- Nishat Tasnim
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Laura De la Vega
- Department of Mechanical Engineering, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Shweta Anil Kumar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Laila Abelseth
- Biomedical Engineering Program, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Matthew Alonzo
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Meitham Amereh
- Faculty of Engineering, University of British Columbia-Okanagan Campus, Kelowna, Canada
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
| | - Stephanie M. Willerth
- Biomedical Engineering Program, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
- International Collaboration on Repair Discoveries, University of British Columbia, 818 West 10th Avenue, Vancouver, BC V5Z 1M9 Canada
| |
Collapse
|
32
|
Ye K, Kaplan DL, Bao G, Bettinger C, Forgacs G, Dong C, Khademhosseini A, Ke Y, Leong K, Sambanis A, Sun W, Yin P. Advanced Cell and Tissue Biomanufacturing. ACS Biomater Sci Eng 2018; 4:2292-2307. [PMID: 33435095 DOI: 10.1021/acsbiomaterials.8b00650] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This position paper assesses state-of-the-art advanced biomanufacturing and identifies paths forward to advance this emerging field in biotechnology and biomedical engineering, including new research opportunities and translational and corporate activities. The vision for the field is to see advanced biomanufacturing emerge as a discipline in academic and industrial communities as well as a technological opportunity to spur research and industry growth. To navigate this vision, the paths to move forward and to identify major barriers were a focal point of discussions at a National Science Foundation-sponsored workshop focused on the topic. Some of the major needs include but are not limited to the integration of specific scientific and engineering disciplines and guidance from regulatory agencies, infrastructure requirements, and strategies for reliable systems integration. Some of the recommendations, major targets, and opportunities were also outlined, including some "grand challenges" to spur interest and progress in the field based on the participants at the workshop. Many of these recommendations have been expanded, materialized, and adopted by the field. For instance, the formation of an initial collaboration network in the community was established. This report provides suggestions for the opportunities and challenges to help move the field of advanced biomanufacturing forward. The field is in the early stages of effecting science and technology in biomanufacturing with a bright and important future impact evident based on the rapid scientific advances in recent years and industry progress.
Collapse
Affiliation(s)
- Kaiming Ye
- Department of Biomedical Engineering, Center of Biomanufacturing for Regenerative Medicine, Watson School of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, United States
| | - David L Kaplan
- Department of Biomedical Engineering, School of Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Gang Bao
- Department of Bioengineering, School of Engineering, Rice University, Houston, Texas 77005, United States
| | - Christopher Bettinger
- Department of Materials Science and Engineering, College of Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Gabor Forgacs
- Department of Bioengineering, College of Engineering, University of Missouri, Columbia, Missouri 65211, United States.,Modern Meadow, Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Cheng Dong
- Department of Biomedical Engineering, College of Engineering, Penn State University, University Park, Pennsylvania 16802, United States
| | - Ali Khademhosseini
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| | - Yonggang Ke
- Department of Biomedical Engineering, College of Engineering, Georgia Tech, Atlanta, Georgia 30332, United States
| | - Kam Leong
- Department of Biomedical Engineering, School of Engineering and Applied Science, Columbia University, New York City, New York 10027, United States
| | | | - Wei Sun
- Department of Mechanical Engineering and Mechanics, College of Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States.,Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Peng Yin
- Department of Systems Biology, Harvard Medical School, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
33
|
Challenges in Bio-fabrication of Organoid Cultures. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:53-71. [DOI: 10.1007/5584_2018_216] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
34
|
Baptista LS, Kronemberger GS, Côrtes I, Charelli LE, Matsui RAM, Palhares TN, Sohier J, Rossi AM, Granjeiro JM. Adult Stem Cells Spheroids to Optimize Cell Colonization in Scaffolds for Cartilage and Bone Tissue Engineering. Int J Mol Sci 2018; 19:E1285. [PMID: 29693604 PMCID: PMC5983745 DOI: 10.3390/ijms19051285] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 02/07/2023] Open
Abstract
Top-down tissue engineering aims to produce functional tissues using biomaterials as scaffolds, thus providing cues for cell proliferation and differentiation. Conversely, the bottom-up approach aims to precondition cells to form modular tissues units (building-blocks) represented by spheroids. In spheroid culture, adult stem cells are responsible for their extracellular matrix synthesis, re-creating structures at the tissue level. Spheroids from adult stem cells can be considered as organoids, since stem cells recapitulate differentiation pathways and also represent a promising approach for identifying new molecular targets (biomarkers) for diagnosis and therapy. Currently, spheroids can be used for scaffold-free (developmental engineering) or scaffold-based approaches. The scaffold promotes better spatial organization of individual spheroids and provides a defined geometry for their 3D assembly in larger and complex tissues. Furthermore, spheroids exhibit potent angiogenic and vasculogenic capacity and serve as efficient vascularization units in porous scaffolds for bone tissue engineering. An automated combinatorial approach that integrates spheroids into scaffolds is starting to be investigated for macro-scale tissue biofabrication.
Collapse
Affiliation(s)
- Leandra Santos Baptista
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Xerém, 25245-390 Duque de Caxias, Rio de Janeiro, Brazil.
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
- Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Campus I, 25071-202 Duque de Caxias, Rio de Janeiro, Brazil.
| | - Gabriela Soares Kronemberger
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Xerém, 25245-390 Duque de Caxias, Rio de Janeiro, Brazil.
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
- Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Campus I, 25071-202 Duque de Caxias, Rio de Janeiro, Brazil.
| | - Isis Côrtes
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Xerém, 25245-390 Duque de Caxias, Rio de Janeiro, Brazil.
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
| | - Letícia Emiliano Charelli
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Xerém, 25245-390 Duque de Caxias, Rio de Janeiro, Brazil.
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
| | - Renata Akemi Morais Matsui
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Xerém, 25245-390 Duque de Caxias, Rio de Janeiro, Brazil.
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
| | - Thiago Nunes Palhares
- Brazilian Center for Physics Research, Xavier Sigaud 150, 22290-180 Urca, Rio de Janeiro, Brazil.
| | - Jerome Sohier
- Laboratory of tissue biology and therapeutic engineering-UMR 5305, CNRS, 69007 Lyon, France.
| | - Alexandre Malta Rossi
- Brazilian Center for Physics Research, Xavier Sigaud 150, 22290-180 Urca, Rio de Janeiro, Brazil.
| | - José Mauro Granjeiro
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), 25250-020 Duque de Caxias, Rio de Janeiro, Brazil.
- Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Campus I, 25071-202 Duque de Caxias, Rio de Janeiro, Brazil.
- Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), 24020-140 Niterói, Brazil.
| |
Collapse
|
35
|
Johnson SC, Smith ZL, Sack BS, Steinberg GD. Tissue Engineering and Conduit Substitution. Urol Clin North Am 2018; 45:133-141. [DOI: 10.1016/j.ucl.2017.09.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
36
|
Garzón I, Serrato D, Roda O, Del Carmen Sánchez-Quevedo M, González-Jaranay M, Moreu G, Nieto-Aguilar R, Alaminos M, Campos A. In vitro Cytokeratin Expression Profiling of Human Oral Mucosa Substitutes Developed by Tissue Engineering. Int J Artif Organs 2018; 32:711-9. [DOI: 10.1177/039139880903201002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In this work we performed a study of cytokeratin (CK) expression profiling on human artificial oral mucosa developed in vitro by tissue engineering at different stages of maturation (from immature to well-developed stages) at the protein and mRNA levels. Human artificial oral mucosa was generated in the laboratory using fibrin-agarose biomaterials. As controls, we used human native normal oral mucosa and embryonic oral tissues. Our results demonstrated that human embryonic oral tissues tended to express CK8 and CK19. In contrast, monolayered bioengineered oral mucosa did not show any CK expression by immunohistochemistry whereas bilayered and multilayered artificial oral mucosa showed several markers of stratified epithelia, but did not express CK10. These results suggest that the CK expression pattern is strongly dependent on the maturation state of the artificial tissues and that the CK expression profile of our model of artificial oral mucosa was partially similar to that of the non-keratinized human adult oral mucosa. However, the expression of CK8 by the artificial oral mucosa suggests that these samples correspond to an early stage of development while kept in vitro.
Collapse
Affiliation(s)
- Ingrid Garzón
- Department of Histology (Tissue Engineering Group), Faculty of Medicine, University of Granada, Granada - Spain
| | - Deyanira Serrato
- Department of Histology (Tissue Engineering Group), Faculty of Medicine, University of Granada, Granada - Spain
| | - Olga Roda
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada - Spain
| | | | - Maximino González-Jaranay
- Department of Stomatology, Faculty of Dentistry, University of Granada, Cartuja Campus, Granada - Spain
| | - Gerardo Moreu
- Department of Stomatology, Faculty of Dentistry, University of Granada, Cartuja Campus, Granada - Spain
| | - Renato Nieto-Aguilar
- Department of Histology (Tissue Engineering Group), Faculty of Medicine, University of Granada, Granada - Spain
| | - Miguel Alaminos
- Department of Histology (Tissue Engineering Group), Faculty of Medicine, University of Granada, Granada - Spain
| | - Antonio Campos
- Department of Histology (Tissue Engineering Group), Faculty of Medicine, University of Granada, Granada - Spain
| |
Collapse
|
37
|
Liu F, Liu C, Chen Q, Ao Q, Tian X, Fan J, Tong H, Wang X. Progress in organ 3D bioprinting. Int J Bioprint 2018; 4:128. [PMID: 33102911 PMCID: PMC7582006 DOI: 10.18063/ijb.v4i1.128] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022] Open
Abstract
Three dimensional (3D) printing is a hot topic in today's scientific, technological and commercial areas. It is recognized as the main field which promotes "the Third Industrial Revolution". Recently, human organ 3D bioprinting has been put forward into equity market as a concept stock and attracted a lot of attention. A large number of outstanding scientists have flung themselves into this field and made some remarkable headways. Nevertheless, organ 3D bioprinting is a sophisticated manufacture procedure which needs profound scientific/technological backgrounds/knowledges to accomplish. Especially, large organ 3D bioprinting encounters enormous difficulties and challenges. One of them is to build implantable branched vascular networks in a predefined 3D construct. At present, organ 3D bioprinting still in its infancy and a great deal of work needs to be done. Here we briefly overview some of the achievements of 3D bioprinting technologies in large organ, such as the bone, liver, heart, cartilage and skin, manufacturing.
Collapse
Affiliation(s)
- Fan Liu
- Department of Tissue Engineering, Center of 3D Printing and Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), Shenyang, China
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Chen Liu
- Department of Tissue Engineering, Center of 3D Printing and Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), Shenyang, China
| | - Qiuhong Chen
- Department of Tissue Engineering, Center of 3D Printing and Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), Shenyang, China
| | - Qiang Ao
- Department of Tissue Engineering, Center of 3D Printing and Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), Shenyang, China
| | - Xiaohong Tian
- Department of Tissue Engineering, Center of 3D Printing and Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), Shenyang, China
| | - Jun Fan
- Department of Tissue Engineering, Center of 3D Printing and Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), Shenyang, China
| | - Hao Tong
- Department of Tissue Engineering, Center of 3D Printing and Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), Shenyang, China
| | - Xiaohong Wang
- Department of Tissue Engineering, Center of 3D Printing and Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), Shenyang, China
- Center of Organ Manufacturing, Department of Mechanical Engineering, Tsinghua University, Beijing, P.R. China
| |
Collapse
|
38
|
Abstract
Three-dimensional (3D) in vitro modeling is increasingly relevant as two-dimensional (2D) cultures have been recognized with limits to recapitulate the complex endogenous conditions in the body. Additionally, fabrication technology is more accessible than ever. Bioprinting, in particular, is an additive manufacturing technique that expands the capabilities of in vitro studies by precisely depositing cells embedded within a 3D biomaterial scaffold that acts as temporary extracellular matrix (ECM). More importantly, bioprinting has vast potential for customization. This allows users to manipulate parameters such as scaffold design, biomaterial selection, and cell types, to create specialized biomimetic 3D systems.The development of a 3D system is important to recapitulate the bone marrow (BM) microenvironment since this particular organ cannot be mimicked with other methods such as organoids. The 3D system can be used to study the interactions between native BM cells and metastatic breast cancer cells (BCCs). Although not perfect, such a system can recapitulate the BM microenvironment. Mesenchymal stem cells (MSCs), a key population within the BM, are known to communicate with BCCs invading the BM and to aid in their transition into dormancy. Dormant BCCs are cycling quiescent and resistant to chemotherapy, which allows them to survive in the BM to resurge even after decades. These persisting BCCs have been identified as the stem cell subset. These BCCs exhibit self-renewal and can be induced to differentiate. More importantly, this BCC subset can initiate tumor formation, exert chemoresistance, and form gap junction with endogenous BM stroma, including MSCs. The bioprinted model detailed in this chapter creates a MSC-BC stem cell coculture system to study intercellular interactions in a model that is more representative of the endogenous 3D microenvironment than conventional 2D cultures. The method can reliably seed primary BM MSCs and BC stem cells within a bioprinted scaffold fabricated from CELLINK Bioink. Since bioprinting is a highly customizable technique, parameters described in this method (i.e., cell-cell ratio, scaffold dimensions) can easily be altered to serve other applications, including studies on hematopoietic regulation.
Collapse
Affiliation(s)
- Caitlyn A Moore
- Division of Hematology/Oncology, Department of Medicine, University of Medicine and Dentistry of New Jersey-Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Niloy N Shah
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Caroline P Smith
- Division of Hematology/Oncology, Department of Medicine, University of Medicine and Dentistry of New Jersey-Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Pranela Rameshwar
- Department of Medicine-Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
39
|
Belair DG, Wolf CJ, Wood C, Ren H, Grindstaff R, Padgett W, Swank A, MacMillan D, Fisher A, Winnik W, Abbott BD. Engineering human cell spheroids to model embryonic tissue fusion in vitro. PLoS One 2017; 12:e0184155. [PMID: 28898253 PMCID: PMC5595299 DOI: 10.1371/journal.pone.0184155] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/19/2017] [Indexed: 01/06/2023] Open
Abstract
Epithelial-mesenchymal interactions drive embryonic fusion events during development, and perturbations of these interactions can result in birth defects. Cleft palate and neural tube defects can result from genetic defects or environmental exposures during development, yet very little is known about the effect of chemical exposures on fusion events during human development because of a lack of relevant and robust human in vitro assays of developmental fusion behavior. Given the etiology and prevalence of cleft palate and the relatively simple architecture and composition of the embryonic palate, we sought to develop a three-dimensional culture system that mimics the embryonic palate and could be used to study fusion behavior in vitro using human cells. We engineered size-controlled human Wharton’s Jelly stromal cell (HWJSC) spheroids and established that 7 days of culture in osteogenesis differentiation medium was sufficient to promote an osteogenic phenotype consistent with embryonic palatal mesenchyme. HWJSC spheroids supported the attachment of human epidermal keratinocyte progenitor cells (HPEKp) on the outer spheroid surface likely through deposition of collagens I and IV, fibronectin, and laminin by mesenchymal spheroids. HWJSC spheroids coated in HPEKp cells exhibited fusion behavior in culture, as indicated by the removal of epithelial cells from the seams between spheroids, that was dependent on epidermal growth factor signaling and fibroblast growth factor signaling in agreement with palate fusion literature. The method described here may broadly apply to the generation of three-dimensional epithelial-mesenchymal co-cultures to study developmental fusion events in a format that is amenable to predictive toxicology applications.
Collapse
Affiliation(s)
- David G. Belair
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Cynthia J. Wolf
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Carmen Wood
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Hongzu Ren
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Rachel Grindstaff
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - William Padgett
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Adam Swank
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Denise MacMillan
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Anna Fisher
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Witold Winnik
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Barbara D. Abbott
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
40
|
Abstract
Bioprinting is becoming a must have capability in tissue engineering research. Key to the growth of the field is the inherent flexibility, which can be used to answer basic scientific questions that can only be addressed under 3D culture conditions, or organ-on-chip systems that could quickly replace underperforming animal models. Almost certainly the most challenging application of bioprinting will be for bottom-up tissue construction, which faces many of the same challenges as scaffold-based tissue engineering. In this review, the current state-of-the-art approaches to 3D bioprinting are discussed in terms of performance and suitability. This is complemented by an overview of hydrogel-based bioinks, with a special emphasis on composite biomaterial systems.
Collapse
Affiliation(s)
- Madeline Burke
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol, BS8 1FD, UK
| | - Benjamin M Carter
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Adam W Perriman
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| |
Collapse
|
41
|
Jessop ZM, Al-Sabah A, Gardiner MD, Combellack E, Hawkins K, Whitaker IS. 3D bioprinting for reconstructive surgery: Principles, applications and challenges. J Plast Reconstr Aesthet Surg 2017; 70:1155-1170. [PMID: 28734756 DOI: 10.1016/j.bjps.2017.06.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/26/2017] [Accepted: 06/01/2017] [Indexed: 12/25/2022]
Abstract
Despite the increasing laboratory research in the growing field of 3D bioprinting, there are few reports of successful translation into surgical practice. This review outlines the principles of 3D bioprinting including software and hardware processes, biocompatible technological platforms and suitable bioinks. The advantages of 3D bioprinting over traditional tissue engineering techniques in assembling cells, biomaterials and biomolecules in a spatially controlled manner to reproduce native tissue macro-, micro- and nanoarchitectures are discussed, together with an overview of current progress in bioprinting tissue types relevant for plastic and reconstructive surgery. If successful, this platform technology has the potential to biomanufacture autologous tissue for reconstruction, obviating the need for donor sites or immunosuppression. The biological, technological and regulatory challenges are highlighted, with strategies to overcome these challenges by using an integrated approach from the fields of engineering, biomaterial science, cell biology and reconstructive microsurgery.
Collapse
Affiliation(s)
- Zita M Jessop
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Science, Swansea University Medical School, Swansea, UK; The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, UK
| | - Ayesha Al-Sabah
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Science, Swansea University Medical School, Swansea, UK
| | | | - Emman Combellack
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Science, Swansea University Medical School, Swansea, UK; The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, UK
| | - Karl Hawkins
- Rheology Research Group, Swansea University Medical School, Swansea, UK
| | - Iain S Whitaker
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Science, Swansea University Medical School, Swansea, UK; The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, UK.
| |
Collapse
|
42
|
Ji S, Guvendiren M. Recent Advances in Bioink Design for 3D Bioprinting of Tissues and Organs. Front Bioeng Biotechnol 2017; 5:23. [PMID: 28424770 PMCID: PMC5380738 DOI: 10.3389/fbioe.2017.00023] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/21/2017] [Indexed: 12/26/2022] Open
Abstract
There is a growing demand for alternative fabrication approaches to develop tissues and organs as conventional techniques are not capable of fabricating constructs with required structural, mechanical, and biological complexity. 3D bioprinting offers great potential to fabricate highly complex constructs with precise control of structure, mechanics, and biological matter [i.e., cells and extracellular matrix (ECM) components]. 3D bioprinting is an additive manufacturing approach that utilizes a "bioink" to fabricate devices and scaffolds in a layer-by-layer manner. 3D bioprinting allows printing of a cell suspension into a tissue construct with or without a scaffold support. The most common bioinks are cell-laden hydrogels, decellulerized ECM-based solutions, and cell suspensions. In this mini review, a brief description and comparison of the bioprinting methods, including extrusion-based, droplet-based, and laser-based bioprinting, with particular focus on bioink design requirements are presented. We also present the current state of the art in bioink design including the challenges and future directions.
Collapse
Affiliation(s)
- Shen Ji
- Instructive Biomaterials and Additive Manufacturing (IBAM) Laboratory, Otto H. York Department of Chemical Biological and Pharmaceutical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing (IBAM) Laboratory, Otto H. York Department of Chemical Biological and Pharmaceutical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| |
Collapse
|
43
|
Nazemroaya F, Soheili ZS, Samiei S, Deezagi A, Ahmadieh H, Davari M, Heidari R, Bagheri A, Darvishalipour-Astaneh S. Induced Retro-Differentiation of Human Retinal Pigment Epithelial Cells on PolyHEMA. J Cell Biochem 2017; 118:3080-3089. [PMID: 28370284 DOI: 10.1002/jcb.26014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 03/27/2017] [Indexed: 11/09/2022]
Abstract
Retinal pigment epithelium (RPE) cells represent a great potential to rescue degenerated cells of the damaged retina. Activation of the virtually plastic properties of RPE cells may aid in recovery of retinal degenerative disorders without the need for entire RPE sheet transplantation. Poly (2-hydroxyethyl methacrylate)(PolyHEMA) is one of the most important hydrogels in the biomaterials world. This hydrophobic polymer does not normally support attachment of mammalian cells. In the current study we investigated the effect of PolyHEMA as a cell culture substrate on the growth, differentiation, and plasticity of hRPE cells. hRPE cells were isolated from neonatal human globes and cultured on PolyHEMA and polystyrene substrates (as controls) in 24-well culture plates. DMEM/F12 was supplemented with 10% fetal bovine serum (FBS) and/or 30% human amniotic fluid (HAF) for cultured cells on polystyrene and PolyHEMA coated vessels. Morphology, rate of cell proliferation and cell death, MTT assay, immunocytochemistry and Real-Time RT-PCR were performed to investigate the effects of PolyHEMA on the growth and differentiation of cultured hRPE cells. Proliferation rate of the cells that had been cultured on PolyHEMA was reduced; PolyHEMA did not induce cell death in the hRPE cultures. hRPE cells cultured on PolyHEMA formed many giant spheroid colonies. The giant colonies were re-cultured and the presence of retinal progenitor markers and markers of hRPE cells were detected in cell cultures on PolyHEMA. PolyHEMA seems to be promising for both maintenance and de-differentiation of hRPE cells and expansion of the retinal progenitor cells from the cultures that are originated from hRPE cells. J. Cell. Biochem. 118: 3080-3089, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Fatemeh Nazemroaya
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | | | - Shahram Samiei
- Blood Transfusion Research Centre High Institute for Research and Education in Transfusion, Medicine, Tehran, Iran
| | | | - Hamid Ahmadieh
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Malihe Davari
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Razeih Heidari
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Abouzar Bagheri
- Genetic Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | | |
Collapse
|
44
|
Moldovan NI, Hibino N, Nakayama K. Principles of the Kenzan Method for Robotic Cell Spheroid-Based Three-Dimensional Bioprinting<sup/>. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:237-244. [PMID: 27917703 DOI: 10.1089/ten.teb.2016.0322] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Bioprinting is a technology with the prospect to change the way many diseases are treated, by replacing the damaged tissues with live de novo created biosimilar constructs. However, after more than a decade of incubation and many proofs of concept, the field is still in its infancy. The current stagnation is the consequence of its early success: the first bioprinters, and most of those that followed, were modified versions of the three-dimensional printers used in additive manufacturing, redesigned for layer-by-layer dispersion of biomaterials. In all variants (inkjet, microextrusion, or laser assisted), this approach is material ("scaffold") dependent and energy intensive, making it hardly compatible with some of the intended biological applications. Instead, the future of bioprinting may benefit from the use of gentler scaffold-free bioassembling methods. A substantial body of evidence has accumulated, indicating this is possible by use of preformed cell spheroids, which have been assembled in cartilage, bone, and cardiac muscle-like constructs. However, a commercial instrument capable to directly and precisely "print" spheroids has not been available until the invention of the microneedles-based ("Kenzan") spheroid assembling and the launching in Japan of a bioprinter based on this method. This robotic platform laces spheroids into predesigned contiguous structures with micron-level precision, using stainless steel microneedles ("kenzans") as temporary support. These constructs are further cultivated until the spheroids fuse into cellular aggregates and synthesize their own extracellular matrix, thus attaining the needed structural organization and robustness. This novel technology opens wide opportunities for bioengineering of tissues and organs.
Collapse
Affiliation(s)
- Nicanor I Moldovan
- 1 Department of Biomedical Engineering, Schools of Engineering and Medicine, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana.,2 Department of Ophthalmology, Schools of Engineering and Medicine, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| | - Narutoshi Hibino
- 3 Department of Surgery, Division of Cardiac Surgery, Johns Hopkins University , Baltimore, Maryland
| | - Koichi Nakayama
- 4 Department of Regenerative Medicine and Biomedical Engineering, Faculty of Medicine, Saga University , Japan
| |
Collapse
|
45
|
|
46
|
Xin Y, Chai G, Zhang T, Wang X, Qu M, Tan A, Bogari M, Zhu M, Lin L, Hu Q, Liu Y, Zhang Y. Analysis of multiple types of human cells subsequent to bioprinting with electrospraying technology. Biomed Rep 2016; 5:723-730. [PMID: 28105339 DOI: 10.3892/br.2016.790] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/04/2016] [Indexed: 12/29/2022] Open
Abstract
The aim of the present study was to investigate bioprinting with electrospraying technology using multiple types of human cell suspensions as bio-ink, in order to lay the initial foundations for the application of the bioprinting technology in tissue engineering. In the current study, six types of human cells were selected and cultured, including human fibroblasts, human adipose-derived stem cells (hADSCs), human periodontal ligament cells (HPDLCs), adult human retinal pigment epithelial cells (ARPE-19), human umbilical vascular endothelial cells (HUVECs) and human gastric epithelial cell line (GES-1). Each cell type was divided into two groups, the experimental and control group. All the experimental group cells were electrosprayed using an electrospraying printer (voltage, 15 kV; flow rate, 150 µl/min) and collected in a petri dish placed 15 cm away from the needle (needle diameter, 0.5 mm). Subsequently, cell viability was detected by flow cytometry with a Live/Dead Viability kit. In addition, the cell morphological characteristics were observed with a phase-contrast microscope after 6 h of culturing in order to obtain adherent cells, while cell proliferation was analyzed using a Cell Counting Kit-8 assay. The control groups, without printing, were subjected to the same procedures as the experimental groups. The results of the cell viability and proliferation assays indicated a statistically significant difference after printing between the experiments and control groups only for the hADSCs (P<0.05); by contrast, no significant difference was observed in cell viability and proliferation for the other five cell types (P>0.05). In addition, there were no observable differences between all experimental and the control groups at any examined time point in the terms of cell morphological characteristics. In conclusion, bioprinting based on electrospraying technology demonstrated no distinct negative effect on cell vitality, proliferation and morphology in the present study, and thus the application of this novel technology to cell printing may provide a promising method in tissue engineering.
Collapse
Affiliation(s)
- Yu Xin
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China; Shanghai Tissue Engineering Key Laboratory, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Gang Chai
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China; Shanghai Tissue Engineering Key Laboratory, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Ting Zhang
- Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, P.R. China
| | - Xiangsheng Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China; Shanghai Tissue Engineering Key Laboratory, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Miao Qu
- Clinic for Plastic, Hand and Burns Surgery, RWTH Aachen University Hospital, D-52074 Aachen, Germany
| | - Andy Tan
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Melia Bogari
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Ming Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Li Lin
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Qingxi Hu
- Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, P.R. China
| | - Yuanyuan Liu
- Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, P.R. China
| | - Yan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China; Shanghai Tissue Engineering Key Laboratory, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
47
|
Abstract
The goal of tissue engineering is to mitigate the critical shortage of donor organs via in vitro fabrication of functional biological structures. Tissue engineering is one of the most prominent examples of interdisciplinary fields, where scientists with different backgrounds work together to boost the quality of life by addressing critical health issues. Many different fields, such as developmental and molecular biology, as well as technologies, such as micro- and nanotechnologies and additive manufacturing, have been integral for advancing the field of tissue engineering. Over the past 20 years, spectacular advancements have been achieved to harness nature's ability to cure diseased tissues and organs. Patients have received laboratory-grown tissues and organs made out of their own cells, thus eliminating the risk of rejection. However, challenges remain when addressing more complex solid organs such as the heart, liver, and kidney. Herein, we review recent accomplishments as well as challenges that must be addressed in the field of tissue engineering and provide a perspective regarding strategies in further development.
Collapse
Affiliation(s)
- Ashkan Shafiee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157; ,
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157; ,
| |
Collapse
|
48
|
Singh D, Singh D, Han SS. 3D Printing of Scaffold for Cells Delivery: Advances in Skin Tissue Engineering. Polymers (Basel) 2016; 8:polym8010019. [PMID: 30979115 PMCID: PMC6432526 DOI: 10.3390/polym8010019] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 01/08/2016] [Accepted: 01/08/2016] [Indexed: 01/01/2023] Open
Abstract
Injury or damage to tissue and organs is a major health problem, resulting in about half of the world’s annual healthcare expenditure every year. Advances in the fields of stem cells (SCs) and biomaterials processing have provided a tremendous leap for researchers to manipulate the dynamics between these two, and obtain a skin substitute that can completely heal the wounded areas. Although wound healing needs a coordinated interplay between cells, extracellular proteins and growth factors, the most important players in this process are the endogenous SCs, which activate the repair cascade by recruiting cells from different sites. Extra cellular matrix (ECM) proteins are activated by these SCs, which in turn aid in cellular migrations and finally secretion of growth factors that can seal and heal the wounds. The interaction between ECM proteins and SCs helps the skin to sustain the rigors of everyday activity, and in an attempt to attain this level of functionality in artificial three-dimensional (3D) constructs, tissue engineered biomaterials are fabricated using more advanced techniques such as bioprinting and laser assisted printing of the organs. This review provides a concise summary of the most recent advances that have been made in the area of polymer bio-fabrication using 3D bio printing used for encapsulating stem cells for skin regeneration. The focus of this review is to describe, in detail, the role of 3D architecture and arrangement of cells within this system that can heal wounds and aid in skin regeneration.
Collapse
Affiliation(s)
- Deepti Singh
- Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT-06510, CT, USA.
| | - Dolly Singh
- Biomaterials Lab, Department of Nano, Medical & Polymer Materials, College of Engineering, Yeungnam University, 280 Daehak-ko, Gyeongsan, Gyeongsanbukdo 712-749, Korea.
| | - Sung Soo Han
- Biomaterials Lab, Department of Nano, Medical & Polymer Materials, College of Engineering, Yeungnam University, 280 Daehak-ko, Gyeongsan, Gyeongsanbukdo 712-749, Korea.
| |
Collapse
|
49
|
Bioprinting a cardiac valve. Biotechnol Adv 2015; 33:1503-21. [DOI: 10.1016/j.biotechadv.2015.07.006] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 06/30/2015] [Accepted: 07/27/2015] [Indexed: 12/13/2022]
|
50
|
Tandon N, Cimetta E, Taubman A, Kupferstein N, Madaan U, Mighty J, Redenti S, Vunjak-Novakovic G. Biomimetic electrical stimulation platform for neural differentiation of retinal progenitor cells. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2013:5666-9. [PMID: 24111023 DOI: 10.1109/embc.2013.6610836] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Electrical activity is abundant in early retinal development, and electrical stimulation has been shown to modulate embryonic stem cell differentiation towards a neuronal fate. The goal of this study was to simulate in vitro retinal developmental electrical activity to drive changes in mouse retinal progenitor cell (mRPC) gene expression and morphology. We designed a biomimetic electrical stimulation protocol based on spontaneous waves present during retinal development, and applied it to retinal progenitor cells (RPCs) over 3 days of culture. Analysis of protein localization and calcium dynamics, indicate that mRPCs undergo functional neuronal maturation. Our findings suggest that this type of electrical stimulation may be utilized for application in neural tissue engineering and open possibilities for understanding mechanisms guiding active electric membrane development and functional organization during early retinogenesis.
Collapse
|