51
|
Tarassoli SP, Jessop ZM, Jovic T, Hawkins K, Whitaker IS. Candidate Bioinks for Extrusion 3D Bioprinting-A Systematic Review of the Literature. Front Bioeng Biotechnol 2021; 9:616753. [PMID: 34722473 PMCID: PMC8548422 DOI: 10.3389/fbioe.2021.616753] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/19/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose: Bioprinting is becoming an increasingly popular platform technology for engineering a variety of tissue types. Our aim was to identify biomaterials that have been found to be suitable for extrusion 3D bioprinting, outline their biomechanical properties and biocompatibility towards their application for bioprinting specific tissue types. This systematic review provides an in-depth overview of current biomaterials suitable for extrusion to aid bioink selection for specific research purposes and facilitate design of novel tailored bioinks. Methods: A systematic search was performed on EMBASE, PubMed, Scopus and Web of Science databases according to the PRISMA guidelines. References of relevant articles, between December 2006 to January 2018, on candidate bioinks used in extrusion 3D bioprinting were reviewed by two independent investigators against standardised inclusion and exclusion criteria. Data was extracted on bioprinter brand and model, printing technique and specifications (speed and resolution), bioink material and class of mechanical assessment, cell type, viability, and target tissue. Also noted were authors, study design (in vitro/in vivo), study duration and year of publication. Results: A total of 9,720 studies were identified, 123 of which met inclusion criteria, consisting of a total of 58 reports using natural biomaterials, 26 using synthetic biomaterials and 39 using a combination of biomaterials as bioinks. Alginate (n = 50) and PCL (n = 33) were the most commonly used bioinks, followed by gelatin (n = 18) and methacrylated gelatin (GelMA) (n = 16). Pneumatic extrusion bioprinting techniques were the most common (n = 78), followed by piston (n = 28). The majority of studies focus on the target tissue, most commonly bone and cartilage, and investigate only one bioink rather than assessing a range to identify those with the most promising printability and biocompatibility characteristics. The Bioscaffolder (GeSiM, Germany), 3D Discovery (regenHU, Switzerland), and Bioplotter (EnvisionTEC, Germany) were the most commonly used commercial bioprinters (n = 35 in total), but groups most often opted to create their own in-house devices (n = 20). Many studies also failed to specify whether the mechanical data reflected pre-, during or post-printing, pre- or post-crosslinking and with or without cells. Conclusions: Despite the continued increase in the variety of biocompatible synthetic materials available, there has been a shift change towards using natural rather than synthetic bioinks for extrusion bioprinting, dominated by alginate either alone or in combination with other biomaterials. On qualitative analysis, no link was demonstrated between the type of bioink or extrusion technique and the target tissue, indicating that bioprinting research is in its infancy with no established tissue specific bioinks or bioprinting techniques. Further research is needed on side-by-side characterisation of bioinks with standardisation of the type and timing of biomechanical assessment.
Collapse
Affiliation(s)
- Sam P Tarassoli
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom
| | - Zita M Jessop
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom.,The Welsh Centre for Burns & Plastic Surgery, Morriston Hospital, Swansea, United Kingdom
| | - Thomas Jovic
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom.,The Welsh Centre for Burns & Plastic Surgery, Morriston Hospital, Swansea, United Kingdom
| | - Karl Hawkins
- Centre for NanoHealth, Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom
| | - Iain S Whitaker
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom.,The Welsh Centre for Burns & Plastic Surgery, Morriston Hospital, Swansea, United Kingdom
| |
Collapse
|
52
|
Gerdes S, Ramesh S, Mostafavi A, Tamayol A, Rivero IV, Rao P. Extrusion-based 3D (Bio)Printed Tissue Engineering Scaffolds: Process-Structure-Quality Relationships. ACS Biomater Sci Eng 2021; 7:4694-4717. [PMID: 34498461 DOI: 10.1021/acsbiomaterials.1c00598] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biological additive manufacturing (Bio-AM) has emerged as a promising approach for the fabrication of biological scaffolds with nano- to microscale resolutions and biomimetic architectures beneficial to tissue engineering applications. However, Bio-AM processes tend to introduce flaws in the construct during fabrication. These flaws can be traced to material nonhomogeneity, suboptimal processing parameters, changes in the (bio)printing environment (such as nozzle clogs), and poor construct design, all with significant contributions to the alteration of a scaffold's mechanical properties. In addition, the biological response of endogenous and exogenous cells interacting with the defective scaffolds could become unpredictable. In this review, we first described extrusion-based Bio-AM. We highlighted the salient architectural and mechanotransduction parameters affecting the response of cells interfaced with the scaffolds. The process phenomena leading to defect formation and some of the tools for defect detection are reviewed. The limitations of the existing developments and the directions that the field should grow in order to overcome said limitations are discussed.
Collapse
Affiliation(s)
- Samuel Gerdes
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0526, United States
| | - Srikanthan Ramesh
- Department of Industrial and Systems Engineering, Rochester Institute of Technology, Rochester, New York. 14623, United States
| | - Azadeh Mostafavi
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0526, United States
| | - Ali Tamayol
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0526, United States.,Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut 06269, United States
| | - Iris V Rivero
- Department of Industrial and Systems Engineering, Rochester Institute of Technology, Rochester, New York. 14623, United States.,Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, New York. 14623, United States
| | - Prahalada Rao
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0526, United States
| |
Collapse
|
53
|
Dellaquila A, Le Bao C, Letourneur D, Simon‐Yarza T. In Vitro Strategies to Vascularize 3D Physiologically Relevant Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100798. [PMID: 34351702 PMCID: PMC8498873 DOI: 10.1002/advs.202100798] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/23/2021] [Indexed: 05/04/2023]
Abstract
Vascularization of 3D models represents a major challenge of tissue engineering and a key prerequisite for their clinical and industrial application. The use of prevascularized models built from dedicated materials could solve some of the actual limitations, such as suboptimal integration of the bioconstructs within the host tissue, and would provide more in vivo-like perfusable tissue and organ-specific platforms. In the last decade, the fabrication of vascularized physiologically relevant 3D constructs has been attempted by numerous tissue engineering strategies, which are classified here in microfluidic technology, 3D coculture models, namely, spheroids and organoids, and biofabrication. In this review, the recent advancements in prevascularization techniques and the increasing use of natural and synthetic materials to build physiological organ-specific models are discussed. Current drawbacks of each technology, future perspectives, and translation of vascularized tissue constructs toward clinics, pharmaceutical field, and industry are also presented. By combining complementary strategies, these models are envisioned to be successfully used for regenerative medicine and drug development in a near future.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Elvesys Microfluidics Innovation CenterParis75011France
- Biomolecular PhotonicsDepartment of PhysicsUniversity of BielefeldBielefeld33615Germany
| | - Chau Le Bao
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Université Sorbonne Paris NordGalilée InstituteVilletaneuseF‐93430France
| | | | | |
Collapse
|
54
|
Abstract
PURPOSE OF REVIEW While liver transplantation is an established treatment for liver failure, the number of patients with liver failure amenable to such intervention far outnumbers the donor supply of livers. Technologies serving to bridge this gap are required. Artificial livers may serve as an alternative. In this review, we discuss the development of artificial liver technologies. RECENT FINDINGS The accrued clinical data suggest that current liver assist devices may serve a role in specific liver diseases, but for the most part no survival benefit has been demonstrated. More clinical trials are expected to elucidate their utilization. Simultaneously, recent advances in materials and tissue engineering are allowing for exciting developments for novel artificial livers. SUMMARY As there continues to be more clinical data regarding the use of current liver devices, new intricate artificial liver technologies, with the use of sophisticated three-dimensional materials, are being developed that may help improve outcomes of liver failure patients.
Collapse
Affiliation(s)
- Asish C Misra
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
55
|
Chen Z, Anandakrishnan N, Xu Y, Zhao R. Compressive Buckling Fabrication of 3D Cell-Laden Microstructures. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101027. [PMID: 34263550 PMCID: PMC8425919 DOI: 10.1002/advs.202101027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/19/2021] [Indexed: 06/13/2023]
Abstract
Tissue architecture is a prerequisite for its biological functions. Recapitulating the three-dimensional (3D) tissue structure represents one of the biggest challenges in tissue engineering. Two-dimensional (2D) tissue fabrication methods are currently in the main stage for tissue engineering and disease modeling. However, due to their planar nature, the created models only represent very limited out-of-plane tissue structure. Here compressive buckling principle is harnessed to create 3D biomimetic cell-laden microstructures from microfabricated planar patterns. This method allows out-of-plane delivery of cells and extracellular matrix patterns with high spatial precision. As a proof of principle, a variety of polymeric 3D miniature structures including a box, an octopus, a pyramid, and continuous waves are fabricated. A mineralized bone tissue model with spatially distributed cell-laden lacunae structures is fabricated to demonstrate the fabrication power of the method. It is expected that this novel approach will help to significantly expand the utility of the established 2D fabrication techniques for 3D tissue fabrication. Given the widespread of 2D fabrication methods in biomedical research and the high demand for biomimetic 3D structures, this method is expected to bridge the gap between 2D and 3D tissue fabrication and open up new possibilities in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Zhaowei Chen
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Nanditha Anandakrishnan
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Ying Xu
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Ruogang Zhao
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, 14260, USA
| |
Collapse
|
56
|
Elalouf A. Immune response against the biomaterials used in 3D bioprinting of organs. Transpl Immunol 2021; 69:101446. [PMID: 34389430 DOI: 10.1016/j.trim.2021.101446] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/26/2022]
Abstract
Regenerative medicine has developed promising approaches for healing and replacing defective and damaged organs or tissues with functional ones. Three-dimensional (3D) bioprinting innovation has integrated a potential to design organs or tissues specific to the patient with the capability of rapid construction to fulfill the storage of organs and the need for transplantation. 3D bioprinting of organs has the main goal to develop a structural and functional organ or tissue mimic to the original one. The highly complex fabrication of tissue engineering scaffolds containing biomaterials, tissue models, and biomedical devices has made it possible to print small blood vessels to mimic organs to reduce organ or tissue rejection. 3D bioprinting has the concept of bioinks containing biomaterials that may trigger the immune responses in the body. Nevertheless, foreign body response (FBR) is mediated by various cell types such as B-cells, dendritic cells, macrophages, natural killer cells, neutrophils, and T-cells, and molecular signals such as antibodies (Abs), cytokines, and reactive radical species. Typically, the biomaterial is shielded by the fibrous encapsulation that is regulated by molecular signals. This review explored the progress in 3D bioprinting of vital organs and basic immune response against the biomaterials used in this approach. Thus, evaluating immune response against biomaterials used in 3D printed organs is necessary to mitigate tissue rejection after the transplantation.
Collapse
Affiliation(s)
- Amir Elalouf
- Bar-Ilan University, Department of Management, Ramat Gan 5290002, Israel.
| |
Collapse
|
57
|
Lam DTUH, Dan YY, Chan YS, Ng HH. Emerging liver organoid platforms and technologies. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:27. [PMID: 34341842 PMCID: PMC8329140 DOI: 10.1186/s13619-021-00089-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/17/2021] [Indexed: 06/13/2023]
Abstract
Building human organs in a dish has been a long term goal of researchers in pursue of physiologically relevant models of human disease and for replacement of worn out and diseased organs. The liver has been an organ of interest for its central role in regulating body homeostasis as well as drug metabolism. An accurate liver replica should contain the multiple cell types found in the organ and these cells should be spatially organized to resemble tissue structures. More importantly, the in vitro model should recapitulate cellular and tissue level functions. Progress in cell culture techniques and bioengineering approaches have greatly accelerated the development of advance 3-dimensional (3D) cellular models commonly referred to as liver organoids. These 3D models described range from single to multiple cell type containing cultures with diverse applications from establishing patient-specific liver cells to modeling of chronic liver diseases and regenerative therapy. Each organoid platform is advantageous for specific applications and presents its own limitations. This review aims to provide a comprehensive summary of major liver organoid platforms and technologies developed for diverse applications.
Collapse
Affiliation(s)
- Do Thuy Uyen Ha Lam
- Laboratory of precision disease therapeutics, Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore
| | - Yock Young Dan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore
- Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Hospital, 5 Lower Kent Ridge Road, Singapore, 119074, Singapore
| | - Yun-Shen Chan
- Laboratory of precision disease therapeutics, Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
| | - Huck-Hui Ng
- Laboratory of precision disease therapeutics, Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore.
- Department of Biochemistry, National University of Singapore, Singapore, 117559, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore.
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117597, Singapore.
| |
Collapse
|
58
|
Khoeini R, Nosrati H, Akbarzadeh A, Eftekhari A, Kavetskyy T, Khalilov R, Ahmadian E, Nasibova A, Datta P, Roshangar L, Deluca DC, Davaran S, Cucchiarini M, Ozbolat IT. Natural and Synthetic Bioinks for 3D Bioprinting. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000097] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Roghayeh Khoeini
- Department of Medicinal Chemistry Faculty of Pharmacy Tabriz University of Medical Sciences P.O. Box: 51664-14766 Tabriz Iran
- Drug Applied Research Center Tabriz University of Medical Sciences P.O. Box: 51656-65811 Tabriz Iran
| | - Hamed Nosrati
- Drug Applied Research Center Tabriz University of Medical Sciences P.O. Box: 51656-65811 Tabriz Iran
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
| | - Abolfazl Akbarzadeh
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Department of Medical Nanotechnology Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences P.O. Box: 516615731 Tabriz Iran
| | - Aziz Eftekhari
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Russian Institute for Advanced Study Moscow State Pedagogical University 1/1, Malaya Pirogovskaya Street Moscow 119991 Russian Federation
- Pharmacology and Toxicology Department Maragheh University of Medical Sciences 78151-55158 Maragheh Iran
- Department of Synthesis and Characterization of Polymers Polymer Institute Slovak Academy of Sciences (SAS) Dúbravská cesta 9 845 41 Bratislava Slovakia
| | - Taras Kavetskyy
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Department of Biology and Chemistry Drohobych Ivan Franko State Pedagogical University 24, I. Franko Str. 82100 Drohobych Ukraine
- Department of Surface Engineering The John Paul II Catholic University of Lublin 20-950 Lublin Poland
| | - Rovshan Khalilov
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Russian Institute for Advanced Study Moscow State Pedagogical University 1/1, Malaya Pirogovskaya Street Moscow 119991 Russian Federation
- Department of Biophysics and Biochemistry Faculty of Biology Baku State University Baku AZ 1143 Azerbaijan
- Institute of Radiation Problems National Academy of Sciences of Azerbaijan Baku AZ 1143 Azerbaijan
| | - Elham Ahmadian
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Kidney Research Center Tabriz University of Medical Sciences P.O. Box: 5166/15731 Tabriz Iran
| | - Aygun Nasibova
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Institute of Radiation Problems National Academy of Sciences of Azerbaijan Baku AZ 1143 Azerbaijan
| | - Pallab Datta
- Department of Pharmaceutics National Institute of Pharmaceutical Education and Research Kolkata West Bengal 700054 India
| | - Leila Roshangar
- Stem Cell Research Center Tabriz University of Medical Sciences P.O. Box: 5166/15731 Tabriz Iran
| | - Dante C. Deluca
- Agricultural and Biological Engineering Department Penn State University University Park 16802 PA USA
| | - Soodabeh Davaran
- Department of Medicinal Chemistry Faculty of Pharmacy Tabriz University of Medical Sciences P.O. Box: 51664-14766 Tabriz Iran
- Drug Applied Research Center Tabriz University of Medical Sciences P.O. Box: 51656-65811 Tabriz Iran
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Department of Medical Nanotechnology Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences P.O. Box: 516615731 Tabriz Iran
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics Saarland University Medical Center Kirrbergerstr. Bldg 37 D-66421 Homburg/Saar Germany
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics Department Penn State University University Park 16802 PA USA
- The Huck Institutes of the Life Sciences Penn State University University Park 16802 PA USA
- Biomedical Engineering Department Penn State University University Park 16802 PA USA
- Materials Research Institute Penn State University University Park 16802 PA USA
- Department of Neurosurgery Penn State University Hershey 17033 PA USA
| |
Collapse
|
59
|
Zhu J, Wang Y, Zhong L, Pan F, Wang J. Advances in tissue engineering of vasculature through three-dimensional bioprinting. Dev Dyn 2021; 250:1717-1738. [PMID: 34115420 DOI: 10.1002/dvdy.385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/07/2021] [Accepted: 06/03/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND A significant challenge facing tissue engineering is the fabrication of vasculature constructs which contains vascularized tissue constructs to recapitulate viable, complex and functional organs or tissues, and free-standing vascular structures potentially providing clinical applications in the future. Three-dimensional (3D) bioprinting has emerged as a promising technology, possessing a number of merits that other conventional biofabrication methods do not have. Over the last decade, 3D bioprinting has contributed a variety of techniques and strategies to generate both vascularized tissue constructs and free-standing vascular structures. RESULTS This review focuses on different strategies to print two kinds of vasculature constructs, namely vascularized tissue constructs and vessel-like tubular structures, highlighting the feasibility and shortcoming of the current methods for vasculature constructs fabrication. Generally, both direct printing and indirect printing can be employed in vascularized tissue engineering. Direct printing allows for structural fabrication with synchronous cell seeding, while indirect printing is more effective in generating complex architecture. During the fabrication process, 3D bioprinting techniques including extrusion bioprinting, inkjet bioprinting and light-assisted bioprinting should be selectively implemented to exert advantages and obtain the desirable tissue structure. Also, appropriate cells and biomaterials matter a lot to match various bioprinting techniques and thus achieve successful fabrication of specific vasculature constructs. CONCLUSION The 3D bioprinting has been developed to help provide various fabrication techniques, devoting to producing structurally stable, physiologically relevant, and biologically appealing constructs. However, although the optimization of biomaterials and innovation of printing strategies may improve the fabricated vessel-like structures, 3D bioprinting is still in the infant period and has a great gap between in vitro trials and in vivo applications. The article reviews the present achievement of 3D bioprinting in generating vasculature constructs and also provides perspectives on future directions of advanced vasculature constructs fabrication.
Collapse
Affiliation(s)
- Junjin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuting Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linna Zhong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fangwei Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
60
|
Esdaille CJ, Washington KS, Laurencin CT. Regenerative engineering: a review of recent advances and future directions. Regen Med 2021; 16:495-512. [PMID: 34030463 PMCID: PMC8356698 DOI: 10.2217/rme-2021-0016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022] Open
Abstract
Regenerative engineering is defined as the convergence of the disciplines of advanced material science, stem cell science, physics, developmental biology and clinical translation for the regeneration of complex tissues and organ systems. It is an expansion of tissue engineering, which was first developed as a method of repair and restoration of human tissue. In the past three decades, advances in regenerative engineering have made it possible to treat a variety of clinical challenges by utilizing cutting-edge technology currently available to harness the body's healing and regenerative abilities. The emergence of new information in developmental biology, stem cell science, advanced material science and nanotechnology have provided promising concepts and approaches to regenerate complex tissues and structures.
Collapse
Affiliation(s)
- Caldon J Esdaille
- Howard University College of Medicine, Washington, DC 20011, USA
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond & Beverly Sackler Center for Biomedical, Biological, Physical & Engineering Sciences, University of Connecticut Health, Farmington, CT 06030, USA
| | - Kenyatta S Washington
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond & Beverly Sackler Center for Biomedical, Biological, Physical & Engineering Sciences, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond & Beverly Sackler Center for Biomedical, Biological, Physical & Engineering Sciences, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06030, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, USA
| |
Collapse
|
61
|
Cui J, Wang HP, Shi Q, Sun T. Pulsed Microfluid Force-Based On-Chip Modular Fabrication for Liver Lobule-Like 3D Cellular Models. CYBORG AND BIONIC SYSTEMS 2021; 2021:9871396. [PMID: 36285127 PMCID: PMC9494728 DOI: 10.34133/2021/9871396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/09/2021] [Indexed: 12/31/2022] Open
Abstract
In vitro three-dimensional (3D) cellular models with native tissue-like architectures and functions have potential as alternatives to human tissues in regenerative medicine and drug discovery. However, it is difficult to replicate liver constructs that mimic in vivo microenvironments using current approaches in tissue engineering because of the vessel-embedded 3D structure and complex cell distribution of the liver. This paper reports a pulsed microflow-based on-chip 3D assembly method to construct 3D liver lobule-like models that replicate the spatial structure and functions of the liver lobule. The heterogeneous cell-laden assembly units with hierarchical cell distribution are fabricated through multistep photopatterning of different cell-laden hydrogels. Through fluid force interaction by pulsed microflow, the hierarchical assembly units are driven to a stack, layer by layer, and thus spatially assemble into 3D cellular models in the closed liquid chamber of the assembly chip. The 3D models with liver lobule-like hexagonal morphology and radial cell distribution allow the dynamic perfusion culture to maintain high cell viability and functional expression during long-term culture in vitro. These results demonstrate that the fabricated 3D liver lobule-like models are promising for drug testing and the study of individual diagnoses and treatments.
Collapse
Affiliation(s)
- J. Cui
- Science and Technology on Electronic Test and Measurement Laboratory, North University of China, Taiyuan 030051, China
- Intelligent Robotics Institute, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - H. P. Wang
- Intelligent Robotics Institute, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Q. Shi
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
| | - T. Sun
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| |
Collapse
|
62
|
Agarwal T, Banerjee D, Konwarh R, Esworthy T, Kumari J, Onesto V, Das P, Lee BH, Wagener FADTG, Makvandi P, Mattoli V, Ghosh SK, Maiti TK, Zhang LG, Ozbolat IT. Recent advances in bioprinting technologies for engineering hepatic tissue. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:112013. [PMID: 33812632 DOI: 10.1016/j.msec.2021.112013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/17/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
In the sphere of liver tissue engineering (LTE), 3D bioprinting has emerged as an effective technology to mimic the complex in vivo hepatic microenvironment, enabling the development of functional 3D constructs with potential application in the healthcare and diagnostic sector. This review gears off with a note on the liver's microscopic 3D architecture and pathologies linked to liver injury. The write-up is then directed towards unmasking recent advancements and prospects of bioprinting for recapitulating 3D hepatic structure and function. The article further introduces available stem cell opportunities and different strategies for their directed differentiation towards various hepatic stem cell types, including hepatocytes, hepatic sinusoidal endothelial cells, stellate cells, and Kupffer cells. Another thrust of the article is on understanding the dynamic interplay of different hepatic cells with various microenvironmental cues, which is crucial for controlling differentiation, maturation, and maintenance of functional hepatic cell phenotype. On a concluding note, various critical issues and future research direction towards clinical translation of bioprinted hepatic constructs are discussed.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Dishary Banerjee
- Department of Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Rocktotpal Konwarh
- Division of Nanobiomaterials and Nanomedicine, Uniglobe Scientific Pvt. Ltd., 7/9, Kishan Garh, Vasant Kunj, New Delhi-110070, India
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Jyoti Kumari
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands; Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), Campus Ecotekne, via Monteroni, Lecce 73100, Italy
| | - Prativa Das
- NTU-Northwestern Institute of Nanomedicine (IGS-NNIN), Nanyang Technological University, 50 Nanyang Ave, Singapore 639798, Singapore
| | - Bae Hoon Lee
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Frank A D T G Wagener
- Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Materials Interface, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Virgilio Mattoli
- Istituto Italiano di Tecnologia, Centre for Materials Interface, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Sudip Kumar Ghosh
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA.
| | - Ibrahim T Ozbolat
- Department of Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA; Materials Research Institute, Penn State University, University Park, PA 16802, USA; Department of Neurosurgery, Penn State College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
63
|
Abstract
Recreating human organ-level function in vitro is a rapidly evolving field that integrates tissue engineering, stem cell biology, and microfluidic technology to produce 3D organoids. A critical component of all organs is the vasculature. Herein, we discuss general strategies to create vascularized organoids, including common source materials, and survey previous work using vascularized organoids to recreate specific organ functions and simulate tumor progression. Vascularization is not only an essential component of individual organ function but also responsible for coupling the fate of all organs and their functions. While some success in coupling two or more organs together on a single platform has been demonstrated, we argue that the future of vascularized organoid technology lies in creating organoid systems complete with tissue-specific microvasculature and in coupling multiple organs through a dynamic vascular network to create systems that can respond to changing physiological conditions.
Collapse
Affiliation(s)
- Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| |
Collapse
|
64
|
Motility Improvement of Biomimetic Trachea Scaffold via Hybrid 3D-Bioprinting Technology. Polymers (Basel) 2021; 13:polym13060971. [PMID: 33810007 PMCID: PMC8004939 DOI: 10.3390/polym13060971] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 11/16/2022] Open
Abstract
A trachea has a structure capable of responding to various movements such as rotation of the neck and relaxation/contraction of the conduit due to the mucous membrane and cartilage tissue. However, current reported tubular implanting structures are difficult to impelement as replacements for original trachea movements. Therefore, in this study, we developed a new trachea implant with similar anatomical structure and mechanical properties to native tissue using 3D printing technology and evaluated its performance. A 250 µm-thick layer composed of polycaprolactone (PCL) nanofibers was fabricated on a rotating beam using electrospinning technology, and a scaffold with C-shaped cartilage grooves that mimics the human airway structure was printed to enable reconstruction of cartilage outside the airway. A cartilage type scaffold had a highest rotational angle (254°) among them and it showed up to 2.8 times compared to human average neck rotation angle. The cartilage type showed a maximum elongation of 8 times higher than that of the bellows type and it showed the elongation of 3 times higher than that of cylinder type. In cartilage type scaffold, gelatin hydrogel printed on the outside of the scaffold was remain 22.2% under the condition where no hydrogel was left in other type scaffolds. In addition, after 2 days of breathing test, the amount of gelatin remaining inside the scaffold was more than twice that of other scaffolds. This novel trachea scaffold with hydrogel inside and outside of the structure was well-preserved under external flow and is expected to be advantageous for soft tissue reconstruction of the trachea.
Collapse
|
65
|
Sun M, Liu A, Yang X, Gong J, Yu M, Yao X, Wang H, He Y. 3D Cell Culture—Can It Be As Popular as 2D Cell Culture? ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Miao Sun
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - An Liu
- Department of Orthopaedic Surgery Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou 310000 China
| | - Xiaofu Yang
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Jiaxing Gong
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Mengfei Yu
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Xinhua Yao
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
| | - Huiming Wang
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Yong He
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
- State Key Laboratory of Fluid Power and Mechatronic Systems School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
| |
Collapse
|
66
|
Ali M, Payne SL. Biomaterial-based cell delivery strategies to promote liver regeneration. Biomater Res 2021; 25:5. [PMID: 33632335 PMCID: PMC7905561 DOI: 10.1186/s40824-021-00206-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/05/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic liver disease and cirrhosis is a widespread and untreatable condition that leads to lifelong impairment and eventual death. The scarcity of liver transplantation options requires the development of new strategies to attenuate disease progression and reestablish liver function by promoting regeneration. Biomaterials are becoming an increasingly promising option to both culture and deliver cells to support in vivo viability and long-term function. There is a wide variety of both natural and synthetic biomaterials that are becoming established as delivery vehicles with their own unique advantages and disadvantages for liver regeneration. We review the latest developments in cell transplantation strategies to promote liver regeneration, with a focus on the use of both natural and synthetic biomaterials for cell culture and delivery. We conclude that future work will need to refine the use of these biomaterials and combine them with novel strategies that recapitulate liver organization and function in order to translate this strategy to clinical use.
Collapse
Affiliation(s)
- Maqsood Ali
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Samantha L Payne
- Department of Biomedical Engineering, School of Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
67
|
Redmond J, McCarthy H, Buchanan P, Levingstone TJ, Dunne NJ. Advances in biofabrication techniques for collagen-based 3D in vitro culture models for breast cancer research. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111944. [PMID: 33641930 DOI: 10.1016/j.msec.2021.111944] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/19/2022]
Abstract
Collagen is the most abundant component of the extracellular matrix (ECM), therefore it represents an ideal biomaterial for the culture of a variety of cell types. Recently, collagen-based scaffolds have shown promise as 3D culture platforms for breast cancer-based research. Two-dimensional (2D) in vitro culture models, while useful for gaining preliminary insights, are ultimately flawed as they do not adequately replicate the tumour microenvironment. As a result, they do not facilitate proper 3D cell-cell/cell-matrix interactions and often an exaggerated response to therapeutic agents occurs. The ECM plays a crucial role in the development and spread of cancer. Alterations within the ECM have a significant impact on the pathogenesis of cancer, the initiation of metastasis and ultimate progression of the disease. 3D in vitro culture models that aim to replicate the tumour microenvironment have the potential to offer a new frontier for cancer research with cell growth, morphology and genetic properties that more closely match in vivo cancers. While initial 3D in vitro culture models used in breast cancer research consisted of simple hydrogel platforms, recent advances in biofabrication techniques, including freeze-drying, electrospinning and 3D bioprinting, have enabled the fabrication of biomimetic collagen-based platforms that more closely replicate the breast cancer ECM. This review highlights the current application of collagen-based scaffolds as 3D in vitro culture models for breast cancer research, specifically for adherence-based scaffolds (i.e. matrix-assisted). Finally, the future perspectives of 3D in vitro breast cancer models and their potential to lead to an improved understanding of breast cancer diagnosis and treatment are discussed.
Collapse
Affiliation(s)
- John Redmond
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland
| | - Helen McCarthy
- School of Pharmacy, Queen's University, Belfast BT9 7BL, United Kingdom; School of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| | - Paul Buchanan
- School of Nursing and Human Science, Dublin City University, Dublin 9, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Tanya J Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
68
|
Abstract
Three-dimensional (3D) printing techniques have revolutionized the field of tissue engineering. This is especially favorable to construct intricate tissues such as liver, as 3D printing allows for the precise delivery of biomaterials, cells and bioactive molecules in complex geometries. Bioinks made of polymers, of both natural and synthetic origin, have been very beneficial to printing soft tissues such as liver. Using polymeric bioinks, 3D hepatic structures are printed with or without cells and biomolecules, and have been used for different tissue engineering applications. In this review, with the introduction to basic 3D printing techniques, we discuss different natural and synthetic polymers including decellularized matrices that have been employed for the 3D bioprinting of hepatic structures. Finally, we focus on recent advances in polymeric bioinks for 3D hepatic printing and their applications. The studies indicate that much work has been devoted to improvising the design, stability and longevity of the printed structures. Others focus on the printing of tissue engineered hepatic structures for applications in drug screening, regenerative medicine and disease models. More attention must now be diverted to developing personalized structures and stem cell differentiation to hepatic lineage.
Collapse
|
69
|
Yu J, Park SA, Kim WD, Ha T, Xin YZ, Lee J, Lee D. Current Advances in 3D Bioprinting Technology and Its Applications for Tissue Engineering. Polymers (Basel) 2020; 12:E2958. [PMID: 33322291 PMCID: PMC7764360 DOI: 10.3390/polym12122958] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/25/2022] Open
Abstract
Three-dimensional (3D) bioprinting technology has emerged as a powerful biofabrication platform for tissue engineering because of its ability to engineer living cells and biomaterial-based 3D objects. Over the last few decades, droplet-based, extrusion-based, and laser-assisted bioprinters have been developed to fulfill certain requirements in terms of resolution, cell viability, cell density, etc. Simultaneously, various bio-inks based on natural-synthetic biomaterials have been developed and applied for successful tissue regeneration. To engineer more realistic artificial tissues/organs, mixtures of bio-inks with various recipes have also been developed. Taken together, this review describes the fundamental characteristics of the existing bioprinters and bio-inks that have been currently developed, followed by their advantages and disadvantages. Finally, various tissue engineering applications using 3D bioprinting are briefly introduced.
Collapse
Affiliation(s)
- JunJie Yu
- Department of Biomedical Engineering, School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul 06974, Korea;
- Department of Nature-Inspired System and Application, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea; (S.A.P.); (W.D.K.)
| | - Su A Park
- Department of Nature-Inspired System and Application, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea; (S.A.P.); (W.D.K.)
| | - Wan Doo Kim
- Department of Nature-Inspired System and Application, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea; (S.A.P.); (W.D.K.)
| | - Taeho Ha
- Department of 3D Printing, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea;
| | - Yuan-Zhu Xin
- Department of Engineering Mechanics, School of Mechanical and Aerospace Engineering, Jilin University, No. 5988, Renmin Street, Changchun 130025, China;
| | - JunHee Lee
- Department of Nature-Inspired System and Application, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea; (S.A.P.); (W.D.K.)
| | - Donghyun Lee
- Department of Biomedical Engineering, School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul 06974, Korea;
| |
Collapse
|
70
|
Ma L, Wu Y, Li Y, Aazmi A, Zhou H, Zhang B, Yang H. Current Advances on 3D-Bioprinted Liver Tissue Models. Adv Healthc Mater 2020; 9:e2001517. [PMID: 33073522 DOI: 10.1002/adhm.202001517] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/27/2020] [Indexed: 12/16/2022]
Abstract
The liver, the largest gland in the human body, plays a key role in metabolism, bile production, detoxification, and water and electrolyte regulation. The toxins or drugs that the gastrointestinal system absorbs reach the liver first before entering the bloodstream. Liver disease is one of the leading causes of death worldwide. Therefore, an in vitro liver tissue model that reproduces the main functions of the liver can be a reliable platform for investigating liver diseases and developing new drugs. In addition, the limitations in traditional, planar monolayer cell cultures and animal tests for evaluating the toxicity and efficacy of drug candidates can be overcome. Currently, the newly emerging 3D bioprinting technologies have the ability to construct in vitro liver tissue models both in static scaffolds and dynamic liver-on-chip manners. This review mainly focuses on the construction and applications of liver tissue models based on 3D bioprinting. Special attention is given to 3D bioprinting strategies and bioinks for constructing liver tissue models including the cell sources and hydrogel selection. In addition, the main advantages and limitations and the major challenges and future perspectives are discussed, paving the way for the next generation of in vitro liver tissue models.
Collapse
Affiliation(s)
- Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou 310027 P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou 310027 P. R. China
| | - Yutong Wu
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou 310027 P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou 310027 P. R. China
| | - Yuting Li
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou 310027 P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou 310027 P. R. China
| | - Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou 310027 P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou 310027 P. R. China
| | - Hongzhao Zhou
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou 310027 P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou 310027 P. R. China
| | - Bin Zhang
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou 310027 P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou 310027 P. R. China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou 310027 P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou 310027 P. R. China
| |
Collapse
|
71
|
Clinical Applications of Patient-Specific 3D Printed Models in Cardiovascular Disease: Current Status and Future Directions. Biomolecules 2020; 10:biom10111577. [PMID: 33233652 PMCID: PMC7699768 DOI: 10.3390/biom10111577] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023] Open
Abstract
Three-dimensional (3D) printing has been increasingly used in medicine with applications in many different fields ranging from orthopaedics and tumours to cardiovascular disease. Realistic 3D models can be printed with different materials to replicate anatomical structures and pathologies with high accuracy. 3D printed models generated from medical imaging data acquired with computed tomography, magnetic resonance imaging or ultrasound augment the understanding of complex anatomy and pathology, assist preoperative planning and simulate surgical or interventional procedures to achieve precision medicine for improvement of treatment outcomes, train young or junior doctors to gain their confidence in patient management and provide medical education to medical students or healthcare professionals as an effective training tool. This article provides an overview of patient-specific 3D printed models with a focus on the applications in cardiovascular disease including: 3D printed models in congenital heart disease, coronary artery disease, pulmonary embolism, aortic aneurysm and aortic dissection, and aortic valvular disease. Clinical value of the patient-specific 3D printed models in these areas is presented based on the current literature, while limitations and future research in 3D printing including bioprinting of cardiovascular disease are highlighted.
Collapse
|
72
|
Chung JJ, Im H, Kim SH, Park JW, Jung Y. Toward Biomimetic Scaffolds for Tissue Engineering: 3D Printing Techniques in Regenerative Medicine. Front Bioeng Biotechnol 2020; 8:586406. [PMID: 33251199 PMCID: PMC7671964 DOI: 10.3389/fbioe.2020.586406] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/05/2020] [Indexed: 12/21/2022] Open
Abstract
Three-dimensional (3D) printing technology allows fabricating complex and precise structures by stacking materials layer by layer. The fabrication method has a strong potential in the regenerative medicine field to produce customizable and defect-fillable scaffolds for tissue regeneration. Plus, biocompatible materials, bioactive molecules, and cells can be printed together or separately to enhance scaffolds, which can save patients who suffer from shortage of transplantable organs. There are various 3D printing techniques that depend on the types of materials, or inks, used. Here, different types of organs (bone, cartilage, heart valve, liver, and skin) that are aided by 3D printed scaffolds and printing methods that are applied in the biomedical fields are reviewed.
Collapse
Affiliation(s)
- Justin J. Chung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, South Korea
| | - Heejung Im
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, South Korea
| | - Soo Hyun Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, South Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| | - Jong Woong Park
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, South Korea
| | - Youngmee Jung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, South Korea
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, South Korea
| |
Collapse
|
73
|
Kim SH, Seo YB, Yeon YK, Lee YJ, Park HS, Sultan MT, Lee JM, Lee JS, Lee OJ, Hong H, Lee H, Ajiteru O, Suh YJ, Song SH, Lee KH, Park CH. 4D-bioprinted silk hydrogels for tissue engineering. Biomaterials 2020; 260:120281. [DOI: 10.1016/j.biomaterials.2020.120281] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 07/13/2020] [Accepted: 08/01/2020] [Indexed: 12/27/2022]
|
74
|
Shah Mohammadi M, Buchen JT, Pasquina PF, Niklason LE, Alvarez LM, Jariwala SH. Critical Considerations for Regeneration of Vascularized Composite Tissues. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:366-381. [PMID: 33115331 DOI: 10.1089/ten.teb.2020.0223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Effective vascularization is vital for survival and functionality of complex tissue-engineered organs. The formation of the microvasculature, composed of endothelial cells (ECs) alone, has been mostly used to restore the vascular networks in organs. However, recent heterocellular studies demonstrate that co-culturing is a more effective approach in revascularization of engineered organs. This review presents key considerations for manufacturing of artificial vascularized composite tissues. We summarize the importance of co-cultures and the multicellular interactions with ECs, as well as design and use of bioreactors, as critical considerations for tissue vascularization. In addition, as an emerging scaffolding technique, this review also highlights the current caveats and hurdles associated with three-dimensional bioprinting and discusses recent developments in bioprinting strategies such as four-dimensional bioprinting and its future outlook for manufacturing of vascularized tissue constructs. Finally, the review concludes with addressing the critical challenges in the regulatory pathway and clinical translation of artificial composite tissue grafts. Impact statement Regeneration of composite tissues is critical as biophysical and biochemical characteristics differ between various types of tissues. Engineering a vascularized composite tissue has remained unresolved and requires additional evaluations along with optimization of methodologies and standard operating procedures. To this end, the main hurdle is creating a viable vascular endothelium that remains functional for a longer duration postimplantation, and can be manufactured using clinically appropriate source of cell lines that are scalable in vitro for the fabrication of human-scale organs. This review presents key considerations for regeneration and manufacturing of vascularized composite tissues as the field advances.
Collapse
Affiliation(s)
- Maziar Shah Mohammadi
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Jack T Buchen
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Paul F Pasquina
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Laura E Niklason
- Department of Anesthesia and Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Luis M Alvarez
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Lung Biotechnology PBC, Silver Spring, Maryland, USA
| | - Shailly H Jariwala
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
75
|
Kim J, Kong JS, Han W, Kim BS, Cho DW. 3D Cell Printing of Tissue/Organ-Mimicking Constructs for Therapeutic and Drug Testing Applications. Int J Mol Sci 2020; 21:E7757. [PMID: 33092184 PMCID: PMC7589604 DOI: 10.3390/ijms21207757] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
The development of artificial tissue/organs with the functional maturity of their native equivalents is one of the long-awaited panaceas for the medical and pharmaceutical industries. Advanced 3D cell-printing technology and various functional bioinks are promising technologies in the field of tissue engineering that have enabled the fabrication of complex 3D living tissue/organs. Various requirements for these tissues, including a complex and large-volume structure, tissue-specific microenvironments, and functional vasculatures, have been addressed to develop engineered tissue/organs with native relevance. Functional tissue/organ constructs have been developed that satisfy such criteria and may facilitate both in vivo replenishment of damaged tissue and the development of reliable in vitro testing platforms for drug development. This review describes key developments in technologies and materials for engineering 3D cell-printed constructs for therapeutic and drug testing applications.
Collapse
Affiliation(s)
- Jongmin Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jeong Sik Kong
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Wonil Han
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Byoung Soo Kim
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
76
|
Lee JM, Suen SKQ, Ng WL, Ma WC, Yeong WY. Bioprinting of Collagen: Considerations, Potentials, and Applications. Macromol Biosci 2020; 21:e2000280. [PMID: 33073537 DOI: 10.1002/mabi.202000280] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/21/2020] [Indexed: 12/15/2022]
Abstract
Collagen is the most abundant extracellular matrix protein that is widely used in tissue engineering (TE). There is little research done on printing pure collagen. To understand the bottlenecks in printing pure collagen, it is imperative to understand collagen from a bottom-up approach. Here it is aimed to provide a comprehensive overview of collagen printing, where collagen assembly in vivo and the various sources of collagen available for TE application are first understood. Next, the current printing technologies and strategy for printing collagen-based materials are highlighted. Considerations and key challenges faced in collagen printing are identified. Finally, the key research areas that would enhance the functionality of printed collagen are presented.
Collapse
Affiliation(s)
- Jia Min Lee
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Sean Kang Qiang Suen
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Wei Long Ng
- HP-NTU Digital Manufacturing Corporate Lab, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Wai Cheung Ma
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Wai Yee Yeong
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore.,HP-NTU Digital Manufacturing Corporate Lab, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
77
|
Abstract
Microvasculature functions at the tissue and cell level, regulating local mass exchange of oxygen and nutrient-rich blood. While there has been considerable success in the biofabrication of large- and small-vessel replacements, functional microvasculature has been particularly challenging to engineer due to its size and complexity. Recently, three-dimensional bioprinting has expanded the possibilities of fabricating sophisticated microvascular systems by enabling precise spatiotemporal placement of cells and biomaterials based on computer-aided design. However, there are still significant challenges facing the development of printable biomaterials that promote robust formation and controlled 3D organization of microvascular networks. This review provides a thorough examination and critical evaluation of contemporary biomaterials and their specific roles in bioprinting microvasculature. We first provide an overview of bioprinting methods and techniques that enable the fabrication of microvessels. We then offer an in-depth critical analysis on the use of hydrogel bioinks for printing microvascularized constructs within the framework of current bioprinting modalities. We end with a review of recent applications of bioprinted microvasculature for disease modeling, drug testing, and tissue engineering, and conclude with an outlook on the challenges facing the evolution of biomaterials design for bioprinting microvasculature with physiological complexity.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael Yost
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
78
|
Fonseca AC, Melchels FPW, Ferreira MJS, Moxon SR, Potjewyd G, Dargaville TR, Kimber SJ, Domingos M. Emulating Human Tissues and Organs: A Bioprinting Perspective Toward Personalized Medicine. Chem Rev 2020; 120:11128-11174. [PMID: 32937071 PMCID: PMC7645917 DOI: 10.1021/acs.chemrev.0c00342] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Indexed: 02/06/2023]
Abstract
The lack of in vitro tissue and organ models capable of mimicking human physiology severely hinders the development and clinical translation of therapies and drugs with higher in vivo efficacy. Bioprinting allow us to fill this gap and generate 3D tissue analogues with complex functional and structural organization through the precise spatial positioning of multiple materials and cells. In this review, we report the latest developments in terms of bioprinting technologies for the manufacturing of cellular constructs with particular emphasis on material extrusion, jetting, and vat photopolymerization. We then describe the different base polymers employed in the formulation of bioinks for bioprinting and examine the strategies used to tailor their properties according to both processability and tissue maturation requirements. By relating function to organization in human development, we examine the potential of pluripotent stem cells in the context of bioprinting toward a new generation of tissue models for personalized medicine. We also highlight the most relevant attempts to engineer artificial models for the study of human organogenesis, disease, and drug screening. Finally, we discuss the most pressing challenges, opportunities, and future prospects in the field of bioprinting for tissue engineering (TE) and regenerative medicine (RM).
Collapse
Affiliation(s)
- Ana Clotilde Fonseca
- Centre
for Mechanical Engineering, Materials and Processes, Department of
Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal
| | - Ferry P. W. Melchels
- Institute
of Biological Chemistry, Biophysics and Bioengineering, School of
Engineering and Physical Sciences, Heriot-Watt
University, Edinburgh EH14 4AS, U.K.
| | - Miguel J. S. Ferreira
- Department
of Mechanical, Aerospace and Civil Engineering, School of Engineering,
Faculty of Science and Engineering, The
University of Manchester, Manchester M13 9PL, U.K.
| | - Samuel R. Moxon
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Geoffrey Potjewyd
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Tim R. Dargaville
- Institute
of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Queensland 4001, Australia
| | - Susan J. Kimber
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Marco Domingos
- Department
of Mechanical, Aerospace and Civil Engineering, School of Engineering,
Faculty of Science and Engineering, The
University of Manchester, Manchester M13 9PL, U.K.
| |
Collapse
|
79
|
Abstract
The field of tissue engineering has advanced over the past decade, but the largest impact on human health should be achieved with the transition of engineered solid organs to the clinic. The number of patients suffering from solid organ disease continues to increase, with over 100 000 patients on the U.S. national waitlist and approximately 730 000 deaths in the United States resulting from end-stage organ disease annually. While flat, tubular, and hollow nontubular engineered organs have already been implanted in patients, in vitro formation of a fully functional solid organ at a translatable scale has not yet been achieved. Thus, one major goal is to bioengineer complex, solid organs for transplantation, composed of patient-specific cells. Among the myriad of approaches attempted to engineer solid organs, 3D bioprinting offers unmatched potential. This review highlights the structural complexity which must be engineered at nano-, micro-, and mesostructural scales to enable organ function. We showcase key advances in bioprinting solid organs with complex vascular networks and functioning microstructures, advances in biomaterials science that have enabled this progress, the regulatory hurdles the field has yet to overcome, and cutting edge technologies that bring us closer to the promise of engineered solid organs.
Collapse
Affiliation(s)
- Adam M Jorgensen
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
80
|
Theus AS, Ning L, Hwang B, Gil C, Chen S, Wombwell A, Mehta R, Serpooshan V. Bioprintability: Physiomechanical and Biological Requirements of Materials for 3D Bioprinting Processes. Polymers (Basel) 2020; 12:E2262. [PMID: 33019639 PMCID: PMC7599870 DOI: 10.3390/polym12102262] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022] Open
Abstract
Three-dimensional (3D) bioprinting is an additive manufacturing process that utilizes various biomaterials that either contain or interact with living cells and biological systems with the goal of fabricating functional tissue or organ mimics, which will be referred to as bioinks. These bioinks are typically hydrogel-based hybrid systems with many specific features and requirements. The characterizing and fine tuning of bioink properties before, during, and after printing are therefore essential in developing reproducible and stable bioprinted constructs. To date, myriad computational methods, mechanical testing, and rheological evaluations have been used to predict, measure, and optimize bioinks properties and their printability, but none are properly standardized. There is a lack of robust universal guidelines in the field for the evaluation and quantification of bioprintability. In this review, we introduced the concept of bioprintability and discussed the significant roles of various physiomechanical and biological processes in bioprinting fidelity. Furthermore, different quantitative and qualitative methodologies used to assess bioprintability will be reviewed, with a focus on the processes related to pre, during, and post printing. Establishing fully characterized, functional bioink solutions would be a big step towards the effective clinical applications of bioprinted products.
Collapse
Affiliation(s)
- Andrea S. Theus
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Boeun Hwang
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Carmen Gil
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Shuai Chen
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Allison Wombwell
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Riya Mehta
- Department of Biology, Emory University, Atlanta, GA 30322, USA;
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
81
|
Bédard P, Gauvin S, Ferland K, Caneparo C, Pellerin È, Chabaud S, Bolduc S. Innovative Human Three-Dimensional Tissue-Engineered Models as an Alternative to Animal Testing. Bioengineering (Basel) 2020; 7:E115. [PMID: 32957528 PMCID: PMC7552665 DOI: 10.3390/bioengineering7030115] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Animal testing has long been used in science to study complex biological phenomena that cannot be investigated using two-dimensional cell cultures in plastic dishes. With time, it appeared that more differences could exist between animal models and even more when translated to human patients. Innovative models became essential to develop more accurate knowledge. Tissue engineering provides some of those models, but it mostly relies on the use of prefabricated scaffolds on which cells are seeded. The self-assembly protocol has recently produced organ-specific human-derived three-dimensional models without the need for exogenous material. This strategy will help to achieve the 3R principles.
Collapse
Affiliation(s)
- Patrick Bédard
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Sara Gauvin
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Karel Ferland
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Christophe Caneparo
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Ève Pellerin
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Stéphane Chabaud
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Stéphane Bolduc
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
82
|
Reinforced gelatin-methacrylate hydrogels containing poly(lactic-co-glycolic acid) nanofiber fragments for 3D bioprinting. J IND ENG CHEM 2020. [DOI: 10.1016/j.jiec.2020.04.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
83
|
Gu Z, Fu J, Lin H, He Y. Development of 3D bioprinting: From printing methods to biomedical applications. Asian J Pharm Sci 2020; 15:529-557. [PMID: 33193859 PMCID: PMC7610207 DOI: 10.1016/j.ajps.2019.11.003] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/22/2019] [Accepted: 11/17/2019] [Indexed: 12/22/2022] Open
Abstract
Biomanufacturing of tissues/organs in vitro is our big dream, driven by two needs: organ transplantation and accurate tissue models. Over the last decades, 3D bioprinting has been widely applied in the construction of many tissues/organs such as skins, vessels, hearts, etc., which can not only lay a foundation for the grand goal of organ replacement, but also be served as in vitro models committed to pharmacokinetics, drug screening and so on. As organs are so complicated, many bioprinting methods are exploited to figure out the challenges of different applications. So the question is how to choose the suitable bioprinting method? Herein, we systematically review the evolution, process and classification of 3D bioprinting with an emphasis on the fundamental printing principles and commercialized bioprinters. We summarize and classify extrusion-based, droplet-based, and photocuring-based bioprinting methods and give some advices for applications. Among them, coaxial and multi-material bioprinting are highlighted and basic principles of designing bioinks are also discussed.
Collapse
Affiliation(s)
- Zeming Gu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jianzhong Fu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Hui Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
84
|
Koyyada A, Orsu P. Recent Advancements and Associated Challenges of Scaffold Fabrication Techniques in Tissue Engineering Applications. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00166-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
85
|
Lee M, Rizzo R, Surman F, Zenobi-Wong M. Guiding Lights: Tissue Bioprinting Using Photoactivated Materials. Chem Rev 2020; 120:10950-11027. [DOI: 10.1021/acs.chemrev.0c00077] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Mihyun Lee
- Tissue Engineering + Biofabrication HPL J22, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Riccardo Rizzo
- Tissue Engineering + Biofabrication HPL J22, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - František Surman
- Tissue Engineering + Biofabrication HPL J22, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication HPL J22, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| |
Collapse
|
86
|
Ramadan Q, Zourob M. Organ-on-a-chip engineering: Toward bridging the gap between lab and industry. BIOMICROFLUIDICS 2020; 14:041501. [PMID: 32699563 PMCID: PMC7367691 DOI: 10.1063/5.0011583] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/22/2020] [Indexed: 05/03/2023]
Abstract
Organ-on-a-chip (OOC) is a very ambitious emerging technology with a high potential to revolutionize many medical and industrial sectors, particularly in preclinical-to-clinical translation in the pharmaceutical arena. In vivo, the function of the organ(s) is orchestrated by a complex cellular structure and physiochemical factors within the extracellular matrix and secreted by various types of cells. The trend in in vitro modeling is to simplify the complex anatomy of the human organ(s) to the minimal essential cellular structure "micro-anatomy" instead of recapitulating the full cellular milieu that enables studying the absorption, metabolism, as well as the mechanistic investigation of drug compounds in a "systemic manner." However, in order to reflect the human physiology in vitro and hence to be able to bridge the gap between the in vivo and in vitro data, simplification should not compromise the physiological relevance. Engineering principles have long been applied to solve medical challenges, and at this stage of organ-on-a-chip technology development, the work of biomedical engineers, focusing on device engineering, is more important than ever to accelerate the technology transfer from the academic lab bench to specialized product development institutions and to the increasingly demanding market. In this paper, instead of presenting a narrative review of the literature, we systemically present a synthesis of the best available organ-on-a-chip technology from what is found, what has been achieved, and what yet needs to be done. We emphasized mainly on the requirements of a "good in vitro model that meets the industrial need" in terms of the structure (micro-anatomy), functions (micro-physiology), and characteristics of the device that hosts the biological model. Finally, we discuss the biological model-device integration supported by an example and the major challenges that delay the OOC technology transfer to the industry and recommended possible options to realize a functional organ-on-a-chip system.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| | - Mohammed Zourob
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| |
Collapse
|
87
|
Firouzian KF, Song Y, Lin F, Zhang T. Fabrication of a biomimetic spinal cord tissue construct with heterogenous mechanical properties using intrascaffold cell assembly. Biotechnol Bioeng 2020; 117:3094-3107. [PMID: 32542651 DOI: 10.1002/bit.27459] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 12/27/2022]
Abstract
In tissue engineering studies, scaffolds play a very important role in offering both physical and chemical cues for cell growth and tissue regeneration. However, in some cases, tissue regeneration requires scaffolds with high mechanical properties (e.g., bone and cartilage), while cells need a soft mechanical microenvironment. In this study, to mimic the heterogenous mechanical properties of a spinal cord tissue, a biomimetic rat tissue construct is fabricated. A collagen-coated poly(lactic-co-glycolic acid) scaffold is manufactured using thermally induced phase separation casting. Primary rat neural cells (P01 Wistar rat cortex) with soft hydrogels are later printed within the scaffold using an image-guided intrascaffold cell assembly technique. The scaffolds have unidirectional microporous structure with parallel axial macrochannels (260 ± 4 µm in diameter). Scaffolds showed mechanical properties similar to rat spine (ultimate tensile strength: 0.085 MPa, Young's modulus [stretch]: 0.31 MPa). The bioink composed of gelatin/alginate/fibrinogen is precisely printed into the macrochannels and showed mechanical properties suitable for neural cells (Young's modulus [compressive]: 3.814 kPa). Scaffold interface, cell viability, and immunostaining analyses show uniform distribution of stable, healthy, and elongated neural cells and neurites over 14 culture days in vitro. The results demonstrated that this method can serve as a valuable tool to aid manufacturing of tissue constructs requiring heterogenous mechanical properties for complex cell and/or biomolecule assembly.
Collapse
Affiliation(s)
- Kevin F Firouzian
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China.,Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, China
| | - Yu Song
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China.,Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, China
| | - Feng Lin
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China.,Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, China
| | - Ting Zhang
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China.,Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, China
| |
Collapse
|
88
|
Salerno S, Tasselli F, Drioli E, De Bartolo L. Poly(ε-Caprolactone) Hollow Fiber Membranes for the Biofabrication of a Vascularized Human Liver Tissue. MEMBRANES 2020; 10:E112. [PMID: 32471264 PMCID: PMC7344547 DOI: 10.3390/membranes10060112] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/23/2020] [Accepted: 05/26/2020] [Indexed: 01/01/2023]
Abstract
The creation of a liver tissue that recapitulates the micro-architecture and functional complexity of a human organ is still one of the main challenges of liver tissue engineering. Here we report on the development of a 3D vascularized hepatic tissue based on biodegradable hollow fiber (HF) membranes of poly(ε-caprolactone) (PCL) that compartmentalize human hepatocytes on the external surface and between the fibers, and endothelial cells into the fiber lumen. To this purpose, PCL HF membranes were prepared by a dry-jet wet phase inversion spinning technique tailoring the operational parameters in order to obtain fibers with suitable properties. After characterization, the fibers were applied to generate a human vascularized hepatic unit by loading endothelial cells in their inner surface and hepatocytes on the external surface. The unit was connected to a perfusion system, and the morpho-functional behavior was evaluated. The results demonstrated the large integration of endothelial cells with the internal surface of individual PCL fibers forming vascular-like structures, and hepatocytes covered completely the external surface and the space between fibers. The perfused 3D hepatic unit retained its functional activity at high levels up to 18 days. This bottom-up tissue engineering approach represents a rational strategy to create relatively 3D vascularized tissues and organs.
Collapse
Affiliation(s)
- Simona Salerno
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
| | - Franco Tasselli
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
| | - Enrico Drioli
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
- Department of Energy, Engineering, Hanyang University, Seoul 04763, Korea
| | - Loredana De Bartolo
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
| |
Collapse
|
89
|
Development of a heat labile antibiotic eluting 3D printed scaffold for the treatment of osteomyelitis. Sci Rep 2020; 10:7554. [PMID: 32371998 PMCID: PMC7200676 DOI: 10.1038/s41598-020-64573-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 04/02/2020] [Indexed: 12/17/2022] Open
Abstract
In general, osteomyelitis is treated with antibiotics, and in severe cases, the inflammatory bone tissue is removed and substituted with poly (methyl methacrylate) (PMMA) beads containing antibiotics. However, this treatment necessitates re-surgery to remove the inserted PMMA beads. Moreover, rifampicin, a primary heat-sensitive antibiotic used for osteomyelitis, is deemed unsuitable in this strategy. Three-dimensional (3D) printing technology has gained popularity, as it facilitates the production of a patient-customized implantable structure using various biodegradable biomaterials as well as controlling printing temperature. Therefore, in this study, we developed a rifampicin-loaded 3D scaffold for the treatment of osteomyelitis using 3D printing and polycaprolactone (PCL), a biodegradable polymer that can be printed at low temperatures. We successfully fabricated rifampicin-loaded PCL 3D scaffolds connected with all pores using computer-aided design and manufacturing (CAD/CAM) and printed them at a temperature of 60 °C to prevent the loss of the antibacterial activity of rifampicin. The growth inhibitory activity against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus), the representative causative organisms of osteomyelitis, was confirmed. In addition, we optimized the rifampicin-loading capacity that causes no damage to the normal bone tissues in 3D scaffold with toxicity evaluation using human osteoblasts. The rifampicin-releasing 3D scaffold developed herein opens new possibilities of the patient-customized treatment of osteomyelitis.
Collapse
|
90
|
Mao Q, Wang Y, Li Y, Juengpanich S, Li W, Chen M, Yin J, Fu J, Cai X. Fabrication of liver microtissue with liver decellularized extracellular matrix (dECM) bioink by digital light processing (DLP) bioprinting. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 109:110625. [DOI: 10.1016/j.msec.2020.110625] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/16/2019] [Accepted: 01/01/2020] [Indexed: 02/07/2023]
|
91
|
Mirdamadi ES, Kalhori D, Zakeri N, Azarpira N, Solati-Hashjin M. Liver Tissue Engineering as an Emerging Alternative for Liver Disease Treatment. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:145-163. [PMID: 31797731 DOI: 10.1089/ten.teb.2019.0233] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic liver diseases affect thousands of lives throughout the world every year. The shortage of liver donors for transplantation has been the main driving force to employ alternative methods such as liver tissue engineering (LTE) in fabricating a three-dimensional transplantable liver tissue or enhancing cell delivery techniques alleviating the need for liver donors. LTE consists of three components, cells, ECM (extracellular matrix), and signaling molecules, which we discuss the first and second. The three most common cell sources used in LTE are human and animal primary hepatocytes, and stem cells for different applications. Two major categories of ECM are used to mimic the microenvironment of these cells, named scaffolds and microbeads. Scaffolds have been made by numerous methods with a wide range of synthetic and natural biomaterials. Cell encapsulation has also been utilized by many polymeric biomaterials. To investigate their functions, many properties have been discussed in the literature, such as biochemical, geometrical, and mechanical properties, in both of these categories. Overall, LTE shows excellent potential in assisting hepatic disorders. However, some challenges exist that prevent the practical use of it clinically, making LTE an ongoing research subject in the scientific society.
Collapse
Affiliation(s)
- Elnaz Sadat Mirdamadi
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Dianoosh Kalhori
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Nima Zakeri
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehran Solati-Hashjin
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
92
|
Progress in 3D bioprinting technology for tissue/organ regenerative engineering. Biomaterials 2020; 226:119536. [DOI: 10.1016/j.biomaterials.2019.119536] [Citation(s) in RCA: 359] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/25/2019] [Accepted: 10/02/2019] [Indexed: 12/21/2022]
|
93
|
Belleghem SMV, Mahadik B, Snodderly KL, Fisher JP. Overview of Tissue Engineering Concepts and Applications. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00081-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
94
|
Kniebs C, Kreimendahl F, Köpf M, Fischer H, Jockenhoevel S, Thiebes AL. Influence of Different Cell Types and Sources on Pre-Vascularisation in Fibrin and Agarose-Collagen Gels. Organogenesis 2019; 16:14-26. [PMID: 31809643 PMCID: PMC7051161 DOI: 10.1080/15476278.2019.1697597] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Vascularisation is essential for the development of tailored, tissue-engineered organs and tissues due to diffusion limits of nutrients and the lack of the necessary connection to the cardiovascular system. To pre-vascularize, endothelial cells and supporting cells can be embedded in the scaffold to foster an adequate nutrient and oxygen supply after transplantation. This technique is applied for tissue engineering of various tissues, but there have been few studies on the use of different cell types or cells sources. We compare the effect of supporting cells from different sources on vascularisation. Fibrin gels and agarose-collagen hydrogels were used as scaffolds. The supporting cells were primary human dermal fibroblasts (HDFs), human nasal fibroblasts (HNFs), human mesenchymal stem cells from umbilical cord’s Wharton’s jelly (WJ MSCs), adipose-derived MSCs (AD MSCs) and femoral bone marrow-derived MSCs (BM MSCs). The tissue constructs were incubated for 14 days and analyzed by two-photon laser scanning microscopy. Vascularisation was supported by all cell types, forming branched networks of tubular vascular structures in both hydrogels. In general, fibrin gels present a higher angiogenic promoting environment compared to agarose-collagen hydrogels and fibroblasts show a high angiogenic potential in co-culture with endothelial cells. In agarose-collagen hydrogels, vascular structures supported by AD MSCs were comparable to our HDF control in terms of volume, area and length. BM MSCs formed a homogeneous network of smaller structures in both hydrogels. This study provides data toward understanding the pre-vascularisation properties of different supporting cell types and sources for tissue engineering of different organs and tissues.
Collapse
Affiliation(s)
- Caroline Kniebs
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany.,Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, The Netherlands
| | - Franziska Kreimendahl
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany.,Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, The Netherlands
| | - Marius Köpf
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany.,Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, The Netherlands
| | - Anja Lena Thiebes
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany.,Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, The Netherlands
| |
Collapse
|
95
|
A Hepatic Scaffold from Decellularized Liver Tissue: Food for Thought. Biomolecules 2019; 9:biom9120813. [PMID: 31810291 PMCID: PMC6995515 DOI: 10.3390/biom9120813] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
Allogeneic liver transplantation is still deemed the gold standard solution for end-stage organ failure; however, donor organ shortages have led to extended waiting lists for organ transplants. In order to overcome the lack of donors, the development of new therapeutic options is mandatory. In the last several years, organ bioengineering has been extensively explored to provide transplantable tissues or whole organs with the final goal of creating a three-dimensional growth microenvironment mimicking the native structure. It has been frequently reported that an extracellular matrix-based scaffold offers a structural support and important biological molecules that could help cellular proliferation during the recellularization process. The aim of the present review is to underline the recent developments in cell-on-scaffold technology for liver bioengineering, taking into account: (1) biological and synthetic scaffolds; (2) animal and human tissue decellularization; (3) scaffold recellularization; (4) 3D bioprinting; and (5) organoid technology. Future possible clinical applications in regenerative medicine for liver tissue engineering and for drug testing were underlined and dissected.
Collapse
|
96
|
Abstract
Engineering approaches were adopted for liver microsystems to recapitulate cell arrangements and culture microenvironments in vivo for sensitive, high-throughput and biomimetic drug screening. This review introduces liver microsystems in vitro for drug hepatotoxicity, drug-drug interactions, metabolic function and enzyme induction, based on cell micropatterning, hydrogel biofabrication and microfluidic perfusion. The engineered microsystems provide varied microenvironments for cell culture that feature cell coculture with non-parenchymal cells, in a heterogeneous extracellular matrix and under controllable perfusion. The engineering methods described include cell micropatterning with soft lithography and dielectrophoresis, hydrogel biofabrication with photolithography, micromolding and 3D bioprinting, and microfluidic perfusion with endothelial-like structures and gradient generators. We discuss the major challenges and trends of liver microsystems to study drug response in vitro.
Collapse
Affiliation(s)
- Jyong-Huei Lee
- Department of Mechanical Engineering, National Taiwan University, Taipei, Taiwan
| | - Kuan-Lun Ho
- Department of Mechanical Engineering, National Taiwan University, Taipei, Taiwan
| | - Shih-Kang Fan
- Department of Mechanical and Nuclear Engineering, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
97
|
Kryou C, Leva V, Chatzipetrou M, Zergioti I. Bioprinting for Liver Transplantation. Bioengineering (Basel) 2019; 6:E95. [PMID: 31658719 PMCID: PMC6956058 DOI: 10.3390/bioengineering6040095] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 12/14/2022] Open
Abstract
Bioprinting techniques can be used for the in vitro fabrication of functional complex bio-structures. Thus, extensive research is being carried on the use of various techniques for the development of 3D cellular structures. This article focuses on direct writing techniques commonly used for the fabrication of cell structures. Three different types of bioprinting techniques are depicted: Laser-based bioprinting, ink-jet bioprinting and extrusion bioprinting. Further on, a special reference is made to the use of the bioprinting techniques for the fabrication of 2D and 3D liver model structures and liver on chip platforms. The field of liver tissue engineering has been rapidly developed, and a wide range of materials can be used for building novel functional liver structures. The focus on liver is due to its importance as one of the most critical organs on which to test new pharmaceuticals, as it is involved in many metabolic and detoxification processes, and the toxicity of the liver is often the cause of drug rejection.
Collapse
Affiliation(s)
- Christina Kryou
- Department of Physics, National Technical University of Athens, 15780 Zografou, Greece.
| | - Valentina Leva
- Department of Physics, National Technical University of Athens, 15780 Zografou, Greece.
| | | | - Ioanna Zergioti
- Department of Physics, National Technical University of Athens, 15780 Zografou, Greece.
| |
Collapse
|
98
|
Zhou Y, Shen JX, Lauschke VM. Comprehensive Evaluation of Organotypic and Microphysiological Liver Models for Prediction of Drug-Induced Liver Injury. Front Pharmacol 2019; 10:1093. [PMID: 31616302 PMCID: PMC6769037 DOI: 10.3389/fphar.2019.01093] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver injury (DILI) is a major concern for the pharmaceutical industry and constitutes one of the most important reasons for the termination of promising drug development projects. Reliable prediction of DILI liability in preclinical stages is difficult, as current experimental model systems do not accurately reflect the molecular phenotype and functionality of the human liver. As a result, multiple drugs that passed preclinical safety evaluations failed due to liver toxicity in clinical trials or postmarketing stages in recent years. To improve the selection of molecules that are taken forward into the clinics, the development of more predictive in vitro systems that enable high-throughput screening of hepatotoxic liabilities and allow for investigative studies into DILI mechanisms has gained growing interest. Specifically, it became increasingly clear that the choice of cell types and culture method both constitute important parameters that affect the predictive power of test systems. In this review, we present current 3D culture paradigms for hepatotoxicity tests and critically evaluate their utility and performance for DILI prediction. In addition, we highlight possibilities of these emerging platforms for mechanistic evaluations of selected drug candidates and present current research directions towards the further improvement of preclinical liver safety tests. We conclude that organotypic and microphysiological liver systems have provided an important step towards more reliable DILI prediction. Furthermore, we expect that the increasing availability of comprehensive benchmarking studies will facilitate model dissemination that might eventually result in their regulatory acceptance.
Collapse
Affiliation(s)
| | | | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
99
|
Sharma D, Ross D, Wang G, Jia W, Kirkpatrick SJ, Zhao F. Upgrading prevascularization in tissue engineering: A review of strategies for promoting highly organized microvascular network formation. Acta Biomater 2019; 95:112-130. [PMID: 30878450 DOI: 10.1016/j.actbio.2019.03.016] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/20/2019] [Accepted: 03/06/2019] [Indexed: 01/05/2023]
Abstract
Functional and perfusable vascular network formation is critical to ensure the long-term survival and functionality of engineered tissues after their transplantation. Although several vascularization strategies have been reviewed in past, the significance of microvessel organization in three-dimensional (3D) scaffolds has been largely ignored. Advances in high-resolution microscopy and image processing have revealed that the majority of tissues including cardiac, skeletal muscle, bone, and skin contain highly organized microvessels that orient themselves to align with tissue architecture for optimum molecular exchange and functional performance. Here, we review strategies to develop highly organized and mature vascular networks in engineered tissues, with a focus on electromechanical stimulation, surface topography, micro scaffolding, surface-patterning, microfluidics and 3D printing. This review will provide researchers with state of the art approaches to engineer vascularized functional tissues for diverse applications. STATEMENT OF SIGNIFICANCE: Vascularization is one of the critical challenges facing tissue engineering. Recent technological advances have enabled researchers to develop microvascular networks in engineered tissues. Although far from translational applications, current vascularization strategies have shown promising outcomes. This review emphasizes the most recent technological advances and future challenges for developing organized microvascular networks in vitro. The next critical step is to achieve highly perfusable, dense, mature and organized microvascular networks representative of native tissues.
Collapse
|
100
|
Leberfinger AN, Dinda S, Wu Y, Koduru SV, Ozbolat V, Ravnic DJ, Ozbolat IT. Bioprinting functional tissues. Acta Biomater 2019; 95:32-49. [PMID: 30639351 PMCID: PMC6625952 DOI: 10.1016/j.actbio.2019.01.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 12/31/2018] [Accepted: 01/09/2019] [Indexed: 12/23/2022]
Abstract
Despite the numerous lives that have been saved since the first successful procedure in 1954, organ transplant has several shortcomings which prevent it from becoming a more comprehensive solution for medical care than it is today. There is a considerable shortage of organ donors, leading to patient death in many cases. In addition, patients require lifelong immunosuppression to prevent graft rejection postoperatively. With such issues in mind, recent research has focused on possible solutions for the lack of access to donor organs and rejections, with the possibility of using the patient's own cells and tissues for treatment showing enormous potential. Three-dimensional (3D) bioprinting is a rapidly emerging technology, which holds great promise for fabrication of functional tissues and organs. Bioprinting offers the means of utilizing a patient's cells to design and fabricate constructs for replacement of diseased tissues and organs. It enables the precise positioning of cells and biologics in an automated and high throughput manner. Several studies have shown the promise of 3D bioprinting. However, many problems must be overcome before the generation of functional tissues with biologically-relevant scale is possible. Specific focus on the functionality of bioprinted tissues is required prior to clinical translation. In this perspective, this paper discusses the challenges of functionalization of bioprinted tissue under eight dimensions: biomimicry, cell density, vascularization, innervation, heterogeneity, engraftment, mechanics, and tissue-specific function, and strives to inform the reader with directions in bioprinting complex and volumetric tissues. STATEMENT OF SIGNIFICANCE: With thousands of patients dying each year waiting for an organ transplant, bioprinted tissues and organs show the potential to eliminate this ever-increasing organ shortage crisis. However, this potential can only be realized by better understanding the functionality of the organ and developing the ability to translate this to the bioprinting methodologies. Considering the rate at which the field is currently expanding, it is reasonable to expect bioprinting to become an integral component of regenerative medicine. For this purpose, this paper discusses several factors that are critical for printing functional tissues including cell density, vascularization, innervation, heterogeneity, engraftment, mechanics, and tissue-specific function, and inform the reader with future directions in bioprinting complex and volumetric tissues.
Collapse
Affiliation(s)
- Ashley N Leberfinger
- Department of Surgery, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Shantanab Dinda
- Department of Industrial and Manufacturing Engineering, The Pennsylvania State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yang Wu
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Srinivas V Koduru
- Department of Surgery, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Veli Ozbolat
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; Ceyhan Engineering Faculty, Cukurova University, Ceyhan, Adana 01950, Turkey
| | - Dino J Ravnic
- Department of Surgery, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Ibrahim T Ozbolat
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA; Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|