1
|
Ghandehari A, Tavares-Negrete JA, Rajendran J, Yi Q, Esfandyarpour R. Optimization of process parameters in 3D-nanomaterials printing for enhanced uniformity, quality, and dimensional precision using physics-guided artificial neural network. DISCOVER NANO 2024; 19:204. [PMID: 39680278 DOI: 10.1186/s11671-024-04155-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
Pneumatic 3D-nanomaterial printing, a prominent additive manufacturing technique, excels in processing advanced materials like MXene, crucial for applications in nano-energy, flexible electronics, and sensors. A key challenge in this domain is optimizing process parameters-applied pressure, ink concentration, nozzle diameter, and printing velocity-to achieve uniform, high-quality prints with the desired filament diameter. Traditional trial-and-error methods often result in significant material waste and time consumption. To address this, our study introduces a comprehensive pipeline that initially assesses whether the selected process parameters yield uniform, high-quality MXene prints. Subsequently, it employs a Physics-Guided Artificial Neural Network (PGANN) to predict the filament diameter based on these parameters, integrating fundamental physical principles of the printing process with experimental data. Our findings demonstrate that using an XGBoost classifier, we can classify printed filament quality with an accuracy of 90.44%. Furthermore, the PGANN model shows exceptional performance in predicting the filament diameter, achieving a Pearson Correlation Coefficient (PCC) of 0.9488, a Mean Squared Error (MSE) of 0.000092 mm2, and a Mean Absolute Error (MAE) of 0.00711 mm. This pipeline significantly streamlines the process for researchers, facilitating the selection of optimal printing parameters to consistently achieve high-quality prints and accurately produce the desired filament diameter tailored to specific applications.
Collapse
Affiliation(s)
- Anita Ghandehari
- Department of Electrical Engineering and Computer Science, University of California, Irvine, CA, 92697, USA
- Henry Samueli School of Engineering, University of California, Irvine, CA, 92697, USA
- Laboratory for Integrated Nano Bio Electronics Innovation, The Henry Samueli School of Engineering, University of California, Irvine, CA, 92697, USA
| | - Jorge A Tavares-Negrete
- Henry Samueli School of Engineering, University of California, Irvine, CA, 92697, USA
- Laboratory for Integrated Nano Bio Electronics Innovation, The Henry Samueli School of Engineering, University of California, Irvine, CA, 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA
| | - Jerome Rajendran
- Department of Electrical Engineering and Computer Science, University of California, Irvine, CA, 92697, USA
- Henry Samueli School of Engineering, University of California, Irvine, CA, 92697, USA
- Laboratory for Integrated Nano Bio Electronics Innovation, The Henry Samueli School of Engineering, University of California, Irvine, CA, 92697, USA
| | - Qian Yi
- Department of Electrical Engineering and Computer Science, University of California, Irvine, CA, 92697, USA
- Laboratory for Integrated Nano Bio Electronics Innovation, The Henry Samueli School of Engineering, University of California, Irvine, CA, 92697, USA
| | - Rahim Esfandyarpour
- Department of Electrical Engineering and Computer Science, University of California, Irvine, CA, 92697, USA.
- Henry Samueli School of Engineering, University of California, Irvine, CA, 92697, USA.
- Laboratory for Integrated Nano Bio Electronics Innovation, The Henry Samueli School of Engineering, University of California, Irvine, CA, 92697, USA.
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.
- Department of Mechanical and Aerospace Engineering, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
2
|
Chiticaru EA, Ioniță M. Commercially available bioinks and state-of-the-art lab-made formulations for bone tissue engineering: A comprehensive review. Mater Today Bio 2024; 29:101341. [PMID: 39649248 PMCID: PMC11625167 DOI: 10.1016/j.mtbio.2024.101341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 12/10/2024] Open
Abstract
Bioprinting and bioinks are two of the game changers in bone tissue engineering. This review presents different bioprinting technologies including extrusion-based, inkjet-based, laser-assisted, light-based, and hybrid technologies with their own strengths and weaknesses. This review will aid researchers in the selection and assessment of the bioink; the discussion ranges from commercially available bioinks to custom lab-made formulations mainly based on natural polymers, such as agarose, alginate, gelatin, collagen, and chitosan, designed for bone tissue engineering. The review is centered on technological advancements and increasing clinical demand within the rapidly growing bioprinting market. From this point of view, 4D, 5D, and 6D printing technologies promise a future where unprecedented levels of innovation will be involved in fabrication processes leading to more dynamic multifunctionalities of bioprinted constructs. Further advances in bioprinting technology, such as hybrid bioprinting methods are covered, with the promise to meet personalized medicine goals while advancing patient outcomes for bone tissues engineering applications.
Collapse
Affiliation(s)
- Elena Alina Chiticaru
- Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, Gh Polizu 1-7, 011061, Bucharest, Romania
| | - Mariana Ioniță
- Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, Gh Polizu 1-7, 011061, Bucharest, Romania
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, Gh Polizu 1-7, 011061, Bucharest, Romania
| |
Collapse
|
3
|
Masri S, Fauzi MB, Rajab NF, Lee WH, Zainal Abidin DA, Siew EL. In vitro 3D skin culture and its sustainability in toxicology: a narrative review. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:476-499. [PMID: 39359233 DOI: 10.1080/21691401.2024.2407617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
In current toxicological research, 2D cell cultures and animal models are well- accepted and commonly employed methods. However, these approaches have many drawbacks and are distant from the actual environment in human. To embrace this, great efforts have been made to provide alternative methods for non-animal skin models in toxicology studies with the need for more mechanistically informative methods. This review focuses on the current state of knowledge regarding the in vitro 3D skin model methods, with different functional states that correspond to the sustainability in the field of toxicology testing. We discuss existing toxicology testing methods using in vitro 3D skin models which provide a better understanding of the testing requirements that are needed. The challenges and future landscape in using the in vitro 3D skin models in toxicology testing are also discussed. We are confident that the in vitro 3D skin models application may become an important tool in toxicology in the context of risk assessment.
Collapse
Affiliation(s)
- Syafira Masri
- Department of Tissue Engineering and Regenerative Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Mh Busra Fauzi
- Department of Tissue Engineering and Regenerative Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
- Advance Bioactive Materials-Cells (Adv-BioMaC) UKM Research Group, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Nor Fadilah Rajab
- Centre for Health Aging and Wellness, Faculty of Helath Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Wing-Hin Lee
- Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), Perak, Malaysia
| | | | - Ee Ling Siew
- ASASIpintar Unit, Pusat PERMATA@Pintar Negara, Universiti Kebangsaan Malaysia, Bangi, Malaysia
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Dong Y, Zhou X, Ding Y, Luo Y, Zhao H. Advances in tumor microenvironment: Applications and challenges of 3D bioprinting. Biochem Biophys Res Commun 2024; 730:150339. [PMID: 39032359 DOI: 10.1016/j.bbrc.2024.150339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024]
Abstract
The tumor microenvironment (TME) assumes a pivotal role in the treatment of oncological diseases, given its intricate interplay of diverse cellular components and extracellular matrices. This dynamic ecosystem poses a serious challenge to traditional research methods in many ways, such as high research costs, inefficient translation, poor reproducibility, and low modeling success rates. These challenges require the search for more suitable research methods to accurately model the TME, and the emergence of 3D bioprinting technology is transformative and an important complement to these traditional methods to precisely control the distribution of cells, biomolecules, and matrix scaffolds within the TME. Leveraging digital design, the technology enables personalized studies with high precision, providing essential experimental flexibility. Serving as a critical bridge between in vitro and in vivo studies, 3D bioprinting facilitates the realistic 3D culturing of cancer cells. This comprehensive article delves into cutting-edge developments in 3D bioprinting, encompassing diverse methodologies, biomaterial choices, and various 3D tumor models. Exploration of current challenges, including limited biomaterial options, printing accuracy constraints, low reproducibility, and ethical considerations, contributes to a nuanced understanding. Despite these challenges, the technology holds immense potential for simulating tumor tissues, propelling personalized medicine, and constructing high-resolution organ models, marking a transformative trajectory in oncological research.
Collapse
Affiliation(s)
- Yingying Dong
- The First School of Climical Medicine of Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xue Zhou
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Yunyi Ding
- Department of Emergency Medicine, The Second Affiliated Hospital of Zhejiang University, School, Hangzhou, 310009, China.
| | - Yichen Luo
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Hong Zhao
- The First School of Climical Medicine of Zhejiang Chinese Medical University, Hangzhou, 310053, China; Department of Breast Surgery, The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310060, China.
| |
Collapse
|
5
|
Lee Y, Min J, Kim S, Park W, Ko J, Jeon NL. Recapitulating the Cancer-Immunity Cycle on a Chip. Adv Healthc Mater 2024:e2401927. [PMID: 39221688 DOI: 10.1002/adhm.202401927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/11/2024] [Indexed: 09/04/2024]
Abstract
The cancer-immunity cycle is a fundamental framework for understanding how the immune system interacts with cancer cells, balancing T cell recognition and elimination of tumors while avoiding autoimmune reactions. Despite advancements in immunotherapy, there remains a critical need to dissect each phase of the cycle, particularly the interactions among the tumor, vasculature, and immune system within the tumor microenvironment (TME). Innovative platforms such as organ-on-a-chip, organoids, and bioprinting within microphysiological systems (MPS) are increasingly utilized to enhance the understanding of these interactions. These systems meticulously replicate crucial aspects of the TME and immune responses, providing robust platforms to study cancer progression, immune evasion, and therapeutic interventions with greater physiological relevance. This review explores the latest advancements in MPS technologies for modeling various stages of the cancer-immune cycle, critically evaluating their applications and limitations in advancing the understanding of cancer-immune dynamics and guiding the development of next-generation immunotherapeutic strategies.
Collapse
Affiliation(s)
- Yujin Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaehong Min
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Solbin Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Wooju Park
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Jihoon Ko
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Mechanical Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Advanced Machines and Design, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Qureator, Inc., San Diego, CA, 92110, USA
| |
Collapse
|
6
|
Figueroa-Milla AE, DeMaria W, Wells D, Jeon O, Alsberg E, Rolle MW. Vascular tissues bioprinted with smooth muscle cell-only bioinks in support baths mimic features of native coronary arteries. Biofabrication 2024; 16:045033. [PMID: 39121893 DOI: 10.1088/1758-5090/ad6d8f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
This study explores the bioprinting of a smooth muscle cell-only bioink into ionically crosslinked oxidized methacrylated alginate (OMA) microgel baths to create self-supporting vascular tissues. The impact of OMA microgel support bath methacrylation degree and cell-only bioink dispensing parameters on tissue formation, remodeling, structure and strength was investigated. We hypothesized that reducing dispensing tip diameter from 27 G (210μm) to 30 G (159μm) for cell-only bioink dispensing would reduce tissue wall thickness and improve the consistency of tissue dimensions while maintaining cell viability. Printing with 30 G tips resulted in decreased mean wall thickness (318.6μm) without compromising mean cell viability (94.8%). Histological analysis of cell-only smooth muscle tissues cultured for 14 d in OMA support baths exhibited decreased wall thickness using 30 G dispensing tips, which correlated with increased collagen deposition and alignment. In addition, a TUNEL assay indicated a decrease in cell death in tissues printed with thinner (30 G) dispensing tips. Mechanical testing demonstrated that tissues printed with a 30 G dispensing tip exhibit an increase in ultimate tensile strength compared to those printed with a 27 G dispensing tip. Overall, these findings highlight the importance of precise control over bioprinting parameters to generate mechanically robust tissues when using cell-only bioinks dispensed and cultured within hydrogel support baths. The ability to control print dimensions using cell-only bioinks may enable bioprinting of more complex soft tissue geometries to generatein vitrotissue models.
Collapse
Affiliation(s)
- Andre E Figueroa-Milla
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
| | - William DeMaria
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
| | - Derrick Wells
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Oju Jeon
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States of America
- Departments of Mechanical & Industrial Engineering, Orthopaedic Surgery, and Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL, United States of America
| | - Marsha W Rolle
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
- The Roux Institute at Northeastern University, Portland, ME, United States of America
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States of America
| |
Collapse
|
7
|
Chen A, Wang W, Mao Z, He Y, Chen S, Liu G, Su J, Feng P, Shi Y, Yan C, Lu J. Multimaterial 3D and 4D Bioprinting of Heterogenous Constructs for Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307686. [PMID: 37737521 DOI: 10.1002/adma.202307686] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/06/2023] [Indexed: 09/23/2023]
Abstract
Additive manufacturing (AM), which is based on the principle of layer-by-layer shaping and stacking of discrete materials, has shown significant benefits in the fabrication of complicated implants for tissue engineering (TE). However, many native tissues exhibit anisotropic heterogenous constructs with diverse components and functions. Consequently, the replication of complicated biomimetic constructs using conventional AM processes based on a single material is challenging. Multimaterial 3D and 4D bioprinting (with time as the fourth dimension) has emerged as a promising solution for constructing multifunctional implants with heterogenous constructs that can mimic the host microenvironment better than single-material alternatives. Notably, 4D-printed multimaterial implants with biomimetic heterogenous architectures can provide a time-dependent programmable dynamic microenvironment that can promote cell activity and tissue regeneration in response to external stimuli. This paper first presents the typical design strategies of biomimetic heterogenous constructs in TE applications. Subsequently, the latest processes in the multimaterial 3D and 4D bioprinting of heterogenous tissue constructs are discussed, along with their advantages and challenges. In particular, the potential of multimaterial 4D bioprinting of smart multifunctional tissue constructs is highlighted. Furthermore, this review provides insights into how multimaterial 3D and 4D bioprinting can facilitate the realization of next-generation TE applications.
Collapse
Affiliation(s)
- Annan Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Wanying Wang
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| | - Zhengyi Mao
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Yunhu He
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Shiting Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Guo Liu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Jin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Pei Feng
- State Key Laboratory of High-Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Yusheng Shi
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Chunze Yan
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Jian Lu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Hong Kong Branch of National Precious Metals Material Engineering Research, Center (NPMM), City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
8
|
Rössler L, Nasehi R, Hansen N, Aveic S, Fischer H. Porous collagen scaffolds enable endothelial lumen formation in vitro under both static and dynamic growth conditions. J Biomed Mater Res B Appl Biomater 2024; 112:e35444. [PMID: 38923270 DOI: 10.1002/jbm.b.35444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/26/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Despite recent advances in the field of tissue engineering, the development of complex tissue-like structures in vitro is compromised by the lack of integration of a functioning vasculature. In this study, we propose a mesoscale three-dimensional (3D) in vitro vascularized connective tissue model and demonstrate its feasibility to prompt the self-assembly of endothelial cells into vessel-like structures. Moreover, we investigate the effect of perfusion on the organization of the cells. For this purpose, primary endothelial cells (HUVECs) and a cell line of human foreskin fibroblasts are cultivated in ECM-like matrices made up of freeze-dried collagen scaffolds permeated with collagen type I hydrogel. A tailored bioreactor is designed to investigate the effect of perfusion on self-organization of HUVECs. Immunofluorescent staining, two-photon microscopy, second-harmonic generation imaging, and scanning electron microscopy are applied to visualize the spatial arrangement of the cells. The analyses reveal the formation of hollow, vessel-like structures of HUVECs in hydrogel-permeated collagen scaffolds under both static and dynamic conditions. In conclusion, we demonstrate the feasibility of a 3D porous collagen scaffolding system that enables and maintains the self-organization of HUVECs into vessel-like structures independent of a dynamic flow.
Collapse
Affiliation(s)
- Lena Rössler
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
- Department of Preventive Dentistry, Periodontology and Cariology, University Medical Center, Göttingen, Germany
| | - Ramin Nasehi
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
- DWI - Leibniz Institute for Interactive Materials, Aachen, Germany
| | - Nadja Hansen
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Sanja Aveic
- 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
| |
Collapse
|
9
|
Arriola-Alvarez I, Jaunarena I, Izeta A, Lafuente H. Progenitor Cell Sources for 3D Bioprinting of Lymphatic Vessels and Potential Clinical Application. Tissue Eng Part A 2024; 30:353-366. [PMID: 37950710 DOI: 10.1089/ten.tea.2023.0204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2023] Open
Abstract
The lymphatic system maintains tissue fluid homeostasis and it is involved in the transport of nutrients and immunosurveillance. It also plays a pivotal role in both pathological and regenerative processes. Lymphatic development in the embryo occurs by polarization and proliferation of lymphatic endothelial cells from the lymph sacs, that is, lymphangiogenesis. Alternatively, lymphvasculogenesis further contributes to the formation of lymphatic vessels. In adult tissues, lymphatic formation rarely occurs under physiological conditions, being restricted to pathological processes. In lymphvasculogenesis, progenitor cells seem to be a source of lymphatic vessels. Indeed, mesenchymal stem cells, adipose stem cells, endothelial progenitor cells, and colony-forming endothelial cells are able to promote lymphatic regeneration by different mechanisms, such as direct differentiation and paracrine effects. In this review, we summarize what is known on the diverse stem/progenitor cell niches available for the lymphatic system, emphasizing the potential that these cells hold for lymphatic tissue engineering through 3D bioprinting and their translation to clinical application.
Collapse
Affiliation(s)
- Inazio Arriola-Alvarez
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| | - Ibon Jaunarena
- Gynecology Oncology Unit, Donostia University Hospital, Donostia-San Sebastián, Spain
- Obstetrics and Gynaecology Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- University of the Basque Country (UPV/EHU), Department of Medical Surgical Specialties, Leioa, Spain
| | - Ander Izeta
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- Department of Biomedical Engineering and Sciences, Tecnun-University of Navarra, Donostia-San Sebastián, Spain
| | - Héctor Lafuente
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| |
Collapse
|
10
|
Fritschen A, Lindner N, Scholpp S, Richthof P, Dietz J, Linke P, Guttenberg Z, Blaeser A. High-Scale 3D-Bioprinting Platform for the Automated Production of Vascularized Organs-on-a-Chip. Adv Healthc Mater 2024; 13:e2304028. [PMID: 38511587 PMCID: PMC11469029 DOI: 10.1002/adhm.202304028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/18/2024] [Indexed: 03/22/2024]
Abstract
3D bioprinting possesses the potential to revolutionize contemporary methodologies for fabricating tissue models employed in pharmaceutical research and experimental investigations. This is enhanced by combining bioprinting with advanced organs-on-a-chip (OOCs), which includes a complex arrangement of multiple cell types representing organ-specific cells, connective tissue, and vasculature. However, both OOCs and bioprinting so far demand a high degree of manual intervention, thereby impeding efficiency and inhibiting scalability to meet technological requirements. Through the combination of drop-on-demand bioprinting with robotic handling of microfluidic chips, a print procedure is achieved that is proficient in managing three distinct tissue models on a chip within only a minute, as well as capable of consecutively processing numerous OOCs without manual intervention. This process rests upon the development of a post-printing sealable microfluidic chip, that is compatible with different types of 3D-bioprinters and easily connected to a perfusion system. The capabilities of the automized bioprint process are showcased through the creation of a multicellular and vascularized liver carcinoma model on the chip. The process achieves full vascularization and stable microvascular network formation over 14 days of culture time, with pronounced spheroidal cell growth and albumin secretion of HepG2 serving as a representative cell model.
Collapse
Affiliation(s)
- Anna Fritschen
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Nils Lindner
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Sebastian Scholpp
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Philipp Richthof
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Jonas Dietz
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | | | | | - Andreas Blaeser
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
- Centre for Synthetic BiologyTechnical University of Darmstadt64289DarmstadtGermany
| |
Collapse
|
11
|
Zarate-Sanchez E, George SC, Moya ML, Robertson C. Vascular dysfunction in hemorrhagic viral fevers: opportunities for organotypic modeling. Biofabrication 2024; 16:032008. [PMID: 38749416 PMCID: PMC11151171 DOI: 10.1088/1758-5090/ad4c0b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
The hemorrhagic fever viruses (HFVs) cause severe or fatal infections in humans. Named after their common symptom hemorrhage, these viruses induce significant vascular dysfunction by affecting endothelial cells, altering immunity, and disrupting the clotting system. Despite advances in treatments, such as cytokine blocking therapies, disease modifying treatment for this class of pathogen remains elusive. Improved understanding of the pathogenesis of these infections could provide new avenues to treatment. While animal models and traditional 2D cell cultures have contributed insight into the mechanisms by which these pathogens affect the vasculature, these models fall short in replicatingin vivohuman vascular dynamics. The emergence of microphysiological systems (MPSs) offers promising avenues for modeling these complex interactions. These MPS or 'organ-on-chip' models present opportunities to better mimic human vascular responses and thus aid in treatment development. In this review, we explore the impact of HFV on the vasculature by causing endothelial dysfunction, blood clotting irregularities, and immune dysregulation. We highlight how existing MPS have elucidated features of HFV pathogenesis as well as discuss existing knowledge gaps and the challenges in modeling these interactions using MPS. Understanding the intricate mechanisms of vascular dysfunction caused by HFV is crucial in developing therapies not only for these infections, but also for other vasculotropic conditions like sepsis.
Collapse
Affiliation(s)
- Evelyn Zarate-Sanchez
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Monica L Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- UC Davis Comprehensive Cancer Center, Davis, CA, United States of America
| |
Collapse
|
12
|
Mazari‐Arrighi E, Lépine M, Ayollo D, Faivre L, Larghero J, Chatelain F, Fuchs A. Self-Organization of Long-Lasting Human Endothelial Capillary-Like Networks Guided by DLP Bioprinting. Adv Healthc Mater 2024; 13:e2302830. [PMID: 38366136 PMCID: PMC11468676 DOI: 10.1002/adhm.202302830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/29/2024] [Indexed: 02/18/2024]
Abstract
Tissue engineering holds great promise for regenerative medicine, drug discovery, and as an alternative to animal models. However, as soon as the dimensions of engineered tissue exceed the diffusion limit of oxygen and nutriments, a necrotic core forms leading to irreversible damage. To overcome this constraint, the establishment of a functional perfusion network is essential. In this work, digital light processing bioprinting is used to encapsulate endothelial progenitor cells (EPCs) in 3D light-cured hydrogel scaffolds to guide them toward vascular network formation. In these scaffolds, EPCs proliferate and self-organize within a few days into branched tubular structures with predefined geometry, forming capillary-like vascular tubes or trees of diameters in the range of 10 to 100 µm. Presenting a confluent monolayer wall of cells strongly connect by tight junctions around a central lumen-like space, these structures can be microinjected with a fluorescent dye and are stable for several weeks in vitro. These endothelial structures can be recovered and manipulated in an alginate patch without altering their shape or viability. This approach opens new opportunities for future applications, such as stacking with other cell sheets or multicellular constructs to yield bioengineered tissue with higher complexity and functionality.
Collapse
Affiliation(s)
- Elsa Mazari‐Arrighi
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Matthieu Lépine
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Dmitry Ayollo
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Lionel Faivre
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Jérôme Larghero
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - François Chatelain
- Université de ParisU976 HIPI, InsermParisF‐75006France
- CEAIRIGGrenobleF‐38000France
| | - Alexandra Fuchs
- Université de ParisU976 HIPI, InsermParisF‐75006France
- CEAIRIGGrenobleF‐38000France
| |
Collapse
|
13
|
Sabetkish S, Currie P, Meagher L. Recent trends in 3D bioprinting technology for skeletal muscle regeneration. Acta Biomater 2024; 181:46-66. [PMID: 38697381 DOI: 10.1016/j.actbio.2024.04.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
Skeletal muscle is a pro-regenerative tissue, that utilizes a tissue-resident stem cell system to effect repair upon injury. Despite the demonstrated efficiency of this system in restoring muscle mass after many acute injuries, in conditions of severe trauma such as those evident in volumetric muscle loss (VML) (>20 % by mass), this self-repair capability is unable to restore tissue architecture, requiring interventions which currently are largely surgical. As a possible alternative, the generation of artificial muscle using tissue engineering approaches may also be of importance in the treatment of VML and muscle diseases such as dystrophies. Three-dimensional (3D) bioprinting has been identified as a promising technique for regeneration of the complex architecture of skeletal muscle. This review discusses existing treatment strategies following muscle damage, recent progress in bioprinting techniques, the bioinks used for muscle regeneration, the immunogenicity of scaffold materials, and in vitro and in vivo maturation techniques for 3D bio-printed muscle constructs. The pros and cons of these bioink formulations are also highlighted. Finally, we present the current limitations and challenges in the field and critical factors to consider for bioprinting approaches to become more translationa and to produce clinically relevant engineered muscle. STATEMENT OF SIGNIFICANCE: This review discusses the physiopathology of muscle injuries and existing clinical treatment strategies for muscle damage, the types of bioprinting techniques that have been applied to bioprinting of muscle, and the bioinks commonly used for muscle regeneration. The pros and cons of these bioinks are highlighted. We present a discussion of existing gaps in the literature and critical factors to consider for the translation of bioprinting approaches and to produce clinically relevant engineered muscle. Finally, we provide insights into what we believe will be the next steps required before the realization of the application of tissue-engineered muscle in humans. We believe this manuscript is an insightful, timely, and instructive review that will guide future muscle bioprinting research from a fundamental construct creation approach, down a translational pathway to achieve the desired impact in the clinic.
Collapse
Affiliation(s)
- Shabnam Sabetkish
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Peter Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
14
|
Bosmans C, Ginés Rodriguez N, Karperien M, Malda J, Moreira Teixeira L, Levato R, Leijten J. Towards single-cell bioprinting: micropatterning tools for organ-on-chip development. Trends Biotechnol 2024; 42:739-759. [PMID: 38310021 DOI: 10.1016/j.tibtech.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 02/05/2024]
Abstract
Organs-on-chips (OoCs) hold promise to engineer progressively more human-relevant in vitro models for pharmaceutical purposes. Recent developments have delivered increasingly sophisticated designs, yet OoCs still lack in reproducing the inner tissue physiology required to fully resemble the native human body. This review emphasizes the need to include microarchitectural and microstructural features, and discusses promising avenues to incorporate well-defined microarchitectures down to the single-cell level. We highlight how their integration will significantly contribute to the advancement of the field towards highly organized structural and hierarchical tissues-on-chip. We discuss the combination of state-of-the-art micropatterning technologies to achieve OoCs resembling human-intrinsic complexity. It is anticipated that these innovations will yield significant advances in realization of the next generation of OoC models.
Collapse
Affiliation(s)
- Cécile Bosmans
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Núria Ginés Rodriguez
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Liliana Moreira Teixeira
- Department of Advanced Organ bioengineering and Therapeutics, University of Twente, Enschede, The Netherlands.
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Jeroen Leijten
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
15
|
Janssen R, Benito-Zarza L, Cleijpool P, Valverde MG, Mihăilă SM, Bastiaan-Net S, Garssen J, Willemsen LEM, Masereeuw R. Biofabrication Directions in Recapitulating the Immune System-on-a-Chip. Adv Healthc Mater 2024:e2304569. [PMID: 38625078 DOI: 10.1002/adhm.202304569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Ever since the implementation of microfluidics in the biomedical field, in vitro models have experienced unprecedented progress that has led to a new generation of highly complex miniaturized cell culture platforms, known as Organs-on-a-Chip (OoC). These devices aim to emulate biologically relevant environments, encompassing perfusion and other mechanical and/or biochemical stimuli, to recapitulate key physiological events. While OoCs excel in simulating diverse organ functions, the integration of the immune organs and immune cells, though recent and challenging, is pivotal for a more comprehensive representation of human physiology. This comprehensive review covers the state of the art in the intricate landscape of immune OoC models, shedding light on the pivotal role of biofabrication technologies in bridging the gap between conceptual design and physiological relevance. The multifaceted aspects of immune cell behavior, crosstalk, and immune responses that are aimed to be replicated within microfluidic environments, emphasizing the need for precise biomimicry are explored. Furthermore, the latest breakthroughs and challenges of biofabrication technologies in immune OoC platforms are described, guiding researchers toward a deeper understanding of immune physiology and the development of more accurate and human predictive models for a.o., immune-related disorders, immune development, immune programming, and immune regulation.
Collapse
Affiliation(s)
- Robine Janssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Laura Benito-Zarza
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Pim Cleijpool
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Marta G Valverde
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Silvia M Mihăilă
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, 6708 WG, The Netherlands
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
- Danone Global Research & Innovation Center, Danone Nutricia Research B.V., Utrecht, 3584 CT, The Netherlands
| | - Linette E M Willemsen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| |
Collapse
|
16
|
Zhan Y, Jiang W, Liu Z, Wang Z, Guo K, Sun J. Utilizing bioprinting to engineer spatially organized tissues from the bottom-up. Stem Cell Res Ther 2024; 15:101. [PMID: 38589956 PMCID: PMC11003108 DOI: 10.1186/s13287-024-03712-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 03/31/2024] [Indexed: 04/10/2024] Open
Abstract
In response to the growing demand for organ substitutes, tissue engineering has evolved significantly. However, it is still challenging to create functional tissues and organs. Tissue engineering from the 'bottom-up' is promising on solving this problem due to its ability to construct tissues with physiological complexity. The workflow of this strategy involves two key steps: the creation of building blocks, and the subsequent assembly. There are many techniques developed for the two pivotal steps. Notably, bioprinting is versatile among these techniques and has been widely used in research. With its high level of automation, bioprinting has great capacity in engineering tissues with precision and holds promise to construct multi-material tissues. In this review, we summarize the techniques applied in fabrication and assembly of building blocks. We elaborate mechanisms and applications of bioprinting, particularly in the 'bottom-up' strategy. We state our perspectives on future trends of bottom-up tissue engineering, hoping to provide useful reference for researchers in this field.
Collapse
Affiliation(s)
- Yichen Zhan
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Wenbin Jiang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Zhirong Liu
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China.
| | - Zhenxing Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China.
| | - Ke Guo
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China.
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China.
| |
Collapse
|
17
|
Khiari Z. Recent Developments in Bio-Ink Formulations Using Marine-Derived Biomaterials for Three-Dimensional (3D) Bioprinting. Mar Drugs 2024; 22:134. [PMID: 38535475 PMCID: PMC10971850 DOI: 10.3390/md22030134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 05/01/2024] Open
Abstract
3D bioprinting is a disruptive, computer-aided, and additive manufacturing technology that allows the obtention, layer-by-layer, of 3D complex structures. This technology is believed to offer tremendous opportunities in several fields including biomedical, pharmaceutical, and food industries. Several bioprinting processes and bio-ink materials have emerged recently. However, there is still a pressing need to develop low-cost sustainable bio-ink materials with superior qualities (excellent mechanical, viscoelastic and thermal properties, biocompatibility, and biodegradability). Marine-derived biomaterials, including polysaccharides and proteins, represent a viable and renewable source for bio-ink formulations. Therefore, the focus of this review centers around the use of marine-derived biomaterials in the formulations of bio-ink. It starts with a general overview of 3D bioprinting processes followed by a description of the most commonly used marine-derived biomaterials for 3D bioprinting, with a special attention paid to chitosan, glycosaminoglycans, alginate, carrageenan, collagen, and gelatin. The challenges facing the application of marine-derived biomaterials in 3D bioprinting within the biomedical and pharmaceutical fields along with future directions are also discussed.
Collapse
Affiliation(s)
- Zied Khiari
- National Research Council of Canada, Aquatic and Crop Resource Development Research Centre, 1411 Oxford Street, Halifax, NS B3H 3Z1, Canada
| |
Collapse
|
18
|
Hooper R, Cummings C, Beck A, Vazquez-Armendariz J, Rodriguez C, Dean D. Sheet-based extrusion bioprinting: a new multi-material paradigm providing mid-extrusion micropatterning control for microvascular applications. Biofabrication 2024; 16:025032. [PMID: 38447217 PMCID: PMC10938191 DOI: 10.1088/1758-5090/ad30c8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/17/2024] [Accepted: 03/06/2024] [Indexed: 03/08/2024]
Abstract
As bioprinting advances into clinical relevance with patient-specific tissue and organ constructs, it must be capable of multi-material fabrication at high resolutions to accurately mimick the complex tissue structures found in the body. One of the most fundamental structures to regenerative medicine is microvasculature. Its continuous hierarchical branching vessel networks bridge surgically manipulatable arteries (∼1-6 mm) to capillary beds (∼10µm). Microvascular perfusion must be established quickly for autologous, allogeneic, or tissue engineered grafts to survive implantation and heal in place. However, traditional syringe-based bioprinting techniques have struggled to produce perfusable constructs with hierarchical branching at the resolution of the arterioles (∼100-10µm) found in microvascular tissues. This study introduces the novel CEVIC bioprinting device (i.e.ContinuouslyExtrudedVariableInternalChanneling), a multi-material technology that breaks the current extrusion-based bioprinting paradigm of pushing cell-laden hydrogels through a nozzle as filaments, instead, in the version explored here, extruding thin, wide cell-laden hydrogel sheets. The CEVIC device adapts the chaotic printing approach to control the width and number of microchannels within the construct as it is extruded (i.e. on-the-fly). Utilizing novel flow valve designs, this strategy can produce continuous gradients varying geometry and materials across the construct and hierarchical branching channels with average widths ranging from 621.5 ± 42.92%µm to 11.67 ± 14.99%µm, respectively, encompassing the resolution range of microvascular vessels. These constructs can also include fugitive/sacrificial ink that vacates to leave demonstrably perfusable channels. In a proof-of-concept experiment, a co-culture of two microvascular cell types, endothelial cells and pericytes, sustained over 90% viability throughout 1 week in microchannels within CEVIC-produced gelatin methacryloyl-sodium alginate hydrogel constructs. These results justify further exploration of generating CEVIC-bioprinted microvasculature, such as pre-culturing and implantation studies.
Collapse
Affiliation(s)
- Ryan Hooper
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, United States of America
| | - Caleb Cummings
- Department of Biology, The Ohio State University, Columbus, OH 43210, United States of America
| | - Anna Beck
- Department of Biochemistry, The Ohio State University, Columbus, OH 43210, United States of America
| | - Javier Vazquez-Armendariz
- Department of Materials Science & Engineering, The Ohio State University, Columbus, OH 43210, United States of America
- Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey 64849, NL, Mexico
| | - Ciro Rodriguez
- Department of Materials Science & Engineering, The Ohio State University, Columbus, OH 43210, United States of America
- Laboratorio Nacional de Manufactura Aditiva y Digital (MADiT), Apodaca 66629, NL, Mexico
- Departamento de Ingeniería Mecánica y Materiales Avanzados, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey 64849, NL, Mexico
| | - David Dean
- Department of Materials Science & Engineering, The Ohio State University, Columbus, OH 43210, United States of America
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus, OH 43210, United States of America
| |
Collapse
|
19
|
Qiu B, Wu D, Xue M, Ou L, Zheng Y, Xu F, Jin H, Gao Q, Zhuang J, Cen J, Lin B, Su YC, Chen S, Sun D. 3D Aligned Nanofiber Scaffold Fabrication with Trench-Guided Electrospinning for Cardiac Tissue Engineering. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:4709-4718. [PMID: 38388349 DOI: 10.1021/acs.langmuir.3c03358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Constructing three-dimensional (3D) aligned nanofiber scaffolds is significant for the development of cardiac tissue engineering, which is promising in the field of drug discovery and disease mechanism study. However, the current nanofiber scaffold preparation strategy, which mainly includes manual assembly and hybrid 3D printing, faces the challenge of integrated fabrication of morphology-controllable nanofibers due to its cross-scale structural feature. In this research, a trench-guided electrospinning (ES) strategy was proposed to directly fabricate 3D aligned nanofiber scaffolds with alternative ES and a direct ink writing (DIW) process. The electric field effect of DIW poly(dimethylsiloxane) (PDMS) side walls on guiding whipping ES nanofibers was investigated to construct trench design rules. It was found that the width/height ratio of trenches greatly affected the nanofiber alignment, and the trench width/height ratio of 1.5 provided the nanofiber alignment degree over 60%. As a proof of principle, 3D nanofiber scaffolds with controllable porosity (60-80%) and alignment (30-60%) were fabricated. The effect of the scaffolds was verified by culturing human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), which resulted in the uniform 3D distribution of aligned hiPSC-CMs with ∼1000 μm thickness. Therefore, this printing strategy shows great potential for the efficient engineered tissue construction.
Collapse
Affiliation(s)
- Bin Qiu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen 361102, China
| | - Dongyang Wu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen 361102, China
| | - Mingcheng Xue
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen 361102, China
| | - Lu Ou
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen 361102, China
| | - Yanfei Zheng
- School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Feng Xu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen 361102, China
| | - Hang Jin
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen 361102, China
| | - Qiang Gao
- Guangdong Provincial People's Hospital, Guangzhou 510080, China
| | - Jian Zhuang
- Guangdong Provincial People's Hospital, Guangzhou 510080, China
| | - Jianzheng Cen
- Guangdong Provincial People's Hospital, Guangzhou 510080, China
| | - Bin Lin
- Guangdong Beating Origin Regenerative Medicine Co. Ltd., Foshan 528231, Guangdong, China
| | - Yu-Chuan Su
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 300044, Taiwan, China
| | - Songyue Chen
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen 361102, China
| | - Daoheng Sun
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen 361102, China
| |
Collapse
|
20
|
Guimarães CF, Liu S, Wang J, Purcell E, Ozedirne T, Ren T, Aslan M, Yin Q, Reis RL, Stoyanova T, Demirci U. Co-axial hydrogel spinning for facile biofabrication of prostate cancer-like 3D models. Biofabrication 2024; 16:025017. [PMID: 38306674 DOI: 10.1088/1758-5090/ad2535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 02/01/2024] [Indexed: 02/04/2024]
Abstract
Glandular cancers are amongst the most prevalent types of cancer, which can develop in many different organs, presenting challenges in their detection as well as high treatment variability and failure rates. For that purpose, anticancer drugs are commonly tested in cancer cell lines grown in 2D tissue culture on plastic dishesin vitro, or in animal modelsin vivo. However, 2D culture models diverge significantly from the 3D characteristics of living tissues and animal models require extensive animal use and time. Glandular cancers, such as prostate cancer-the second leading cause of male cancer death-typically exist in co-centrical architectures where a cell layer surrounds an acellular lumen. Herein, this spatial cellular position and 3D architecture, containing dual compartments with different hydrogel materials, is engineered using a simple co-axial nozzle setup, in a single step utilizing prostate as a model of glandular cancer. The resulting hydrogel soft structures support viable prostate cancer cells of different cell lines and enable over-time maturation into cancer-mimicking aggregates surrounding the acellular core. The biofabricated cancer mimicking structures are then used as a model to predict the inhibitory efficacy of the poly ADP ribose polymerase inhibitor, Talazoparib, and the antiandrogen drug, Enzalutamide, in the growth of the cancer cell layer. Our results show that the obtained hydrogel constructs can be adapted to quickly obtain 3D cancer models which combine 3D physiological architectures with high-throughput screening to detect and optimize anti-cancer drugs in prostate and potentially other glandular cancer types.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga and Guimarães, Portugal
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Shiqin Liu
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, United States of America
| | - Jie Wang
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Emma Purcell
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Tugba Ozedirne
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Tanchen Ren
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Merve Aslan
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Qingqing Yin
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Rui L Reis
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga and Guimarães, Portugal
| | - Tanya Stoyanova
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, United States of America
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, United States of America
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| |
Collapse
|
21
|
Zhang Y, O'Mahony A, He Y, Barber T. Hydrodynamic shear stress' impact on mammalian cell properties and its applications in 3D bioprinting. Biofabrication 2024; 16:022003. [PMID: 38277669 DOI: 10.1088/1758-5090/ad22ee] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 01/26/2024] [Indexed: 01/28/2024]
Abstract
As an effective cell assembly method, three-dimensional bioprinting has been widely used in building organ models and tissue repair over the past decade. However, different shear stresses induced throughout the entire printing process can cause complex impacts on cell integrity, including reducing cell viability, provoking morphological changes and altering cellular functionalities. The potential effects that may occur and the conditions under which these effects manifest are not clearly understood. Here, we review systematically how different mammalian cells respond under shear stress. We enumerate available experimental apparatus, and we categorise properties that can be affected under disparate stress patterns. We also summarise cell damaging mathematical models as a predicting reference for the design of bioprinting systems. We concluded that it is essential to quantify specific cell resistance to shear stress for the optimisation of bioprinting systems. Besides, as substantial positive impacts, including inducing cell alignment and promoting cell motility, can be generated by shear stress, we suggest that we find the proper range of shear stress and actively utilise its positive influences in the development of future systems.
Collapse
Affiliation(s)
- Yani Zhang
- School of Mechanical Engineering, UNSW, Sydney, NSW 2052, Australia
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Aidan O'Mahony
- Inventia Life Science Pty Ltd, Alexandria, Sydney, NSW 2015, Australia
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Tracie Barber
- School of Mechanical Engineering, UNSW, Sydney, NSW 2052, Australia
| |
Collapse
|
22
|
Budharaju H, Sundaramurthi D, Sethuraman S. Embedded 3D bioprinting - An emerging strategy to fabricate biomimetic & large vascularized tissue constructs. Bioact Mater 2024; 32:356-384. [PMID: 37920828 PMCID: PMC10618244 DOI: 10.1016/j.bioactmat.2023.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/16/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Three-dimensional bioprinting is an advanced tissue fabrication technique that allows printing complex structures with precise positioning of multiple cell types layer-by-layer. Compared to other bioprinting methods, extrusion bioprinting has several advantages to print large-sized tissue constructs and complex organ models due to large build volume. Extrusion bioprinting using sacrificial, support and embedded strategies have been successfully employed to facilitate printing of complex and hollow structures. Embedded bioprinting is a gel-in-gel approach developed to overcome the gravitational and overhanging limits of bioprinting to print large-sized constructs with a micron-scale resolution. In embedded bioprinting, deposition of bioinks into the microgel or granular support bath will be facilitated by the sol-gel transition of the support bath through needle movement inside the granular medium. This review outlines various embedded bioprinting strategies and the polymers used in the embedded systems with advantages, limitations, and efficacy in the fabrication of complex vascularized tissues or organ models with micron-scale resolution. Further, the essential requirements of support bath systems like viscoelasticity, stability, transparency and easy extraction to print human scale organs are discussed. Additionally, the organs or complex geometries like vascular constructs, heart, bone, octopus and jellyfish models printed using support bath assisted printing methods with their anatomical features are elaborated. Finally, the challenges in clinical translation and the future scope of these embedded bioprinting models to replace the native organs are envisaged.
Collapse
Affiliation(s)
- Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Center for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Center, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Center for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Center, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Center for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Center, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| |
Collapse
|
23
|
Arjoca S, Bojin F, Neagu M, Păunescu A, Neagu A, Păunescu V. Hydrogel Extrusion Speed Measurements for the Optimization of Bioprinting Parameters. Gels 2024; 10:103. [PMID: 38391433 PMCID: PMC10888060 DOI: 10.3390/gels10020103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Three-dimensional (3D) bioprinting is the use of computer-controlled transfer processes for assembling bioinks (cell clusters or materials loaded with cells) into structures of prescribed 3D organization. The correct bioprinting parameters ensure a fast and accurate bioink deposition without exposing the cells to harsh conditions. This study seeks to optimize pneumatic extrusion-based bioprinting based on hydrogel flow rate and extrusion speed measurements. We measured the rate of the hydrogel flow through a cylindrical nozzle and used non-Newtonian hydrodynamics to fit the results. From the videos of free-hanging hydrogel strands delivered from a stationary print head, we inferred the extrusion speed, defined as the speed of advancement of newly formed strands. Then, we relied on volume conservation to evaluate the extrudate swell ratio. The theoretical analysis enabled us to compute the extrusion speed for pressures not tested experimentally as well as the printing speed needed to deposit hydrogel filaments of a given diameter. Finally, the proposed methodology was tested experimentally by analyzing the morphology of triple-layered square-grid hydrogel constructs printed at various applied pressures while the printing speeds matched the corresponding extrusion speeds. Taken together, the results of this study suggest that preliminary measurements and theoretical analyses can simplify the search for the optimal bioprinting parameters.
Collapse
Affiliation(s)
- Stelian Arjoca
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Center for Modeling Biological Systems and Data Analysis, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Florina Bojin
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- OncoGen Institute, 300723 Timisoara, Romania
| | - Monica Neagu
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Center for Modeling Biological Systems and Data Analysis, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Andreea Păunescu
- Carol Davila University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania
| | - Adrian Neagu
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Center for Modeling Biological Systems and Data Analysis, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Department of Physics and Astronomy, University of Missouri, Columbia, MO 65211, USA
| | - Virgil Păunescu
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- OncoGen Institute, 300723 Timisoara, Romania
| |
Collapse
|
24
|
Zhang F, Lin DSY, Rajasekar S, Sotra A, Zhang B. Pump-Less Platform Enables Long-Term Recirculating Perfusion of 3D Printed Tubular Tissues. Adv Healthc Mater 2023; 12:e2300423. [PMID: 37543836 PMCID: PMC11469154 DOI: 10.1002/adhm.202300423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/13/2023] [Indexed: 08/07/2023]
Abstract
The direction and pattern of fluid flow affect vascular structure and function, in which vessel-lining endothelial cells exhibit variable cellular morphologies and vessel remodeling by mechanosensing. To recapitulate this microenvironment, some approaches have been reported to successfully apply unidirectional flow on endothelial cells in organ-on-a-chip systems. However, these platforms encounter drawbacks such as the dependency on pumps or confinement to closed microfluidic channels. These constraints impede their synergy with advanced biofabrication techniques like 3D bioprinting, thereby curtailing the potential to introduce greater complexity into engineered tissues. Herein, a pumpless recirculating platform (UniPlate) that enables unidirectional media recirculation through 3D printed tubular tissues, is demonstrated.The device is made of polystyrene via injection molding in combination with 3D printed sacrifical gelatin templates. Tubular blood vessels with unidirectional perfusion are firstly engineered. Then the design is expanded to incorporate duo-recirculating flow for culturing vascularized renal proximal tubules with glucose reabsorption function. In addition to media recirculation, human monocyte recirculation in engineered blood vessels is also demonstrated for over 24 h, with minimal loss of cells, cell viability, and inflammatory activation. UniPlate can be a valuable tool to more precisely control the cellular microenvironment of organ-on-a-chip systems for drug discovery.
Collapse
Affiliation(s)
- Feng Zhang
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Dawn S. Y. Lin
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | | | - Alexander Sotra
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Boyang Zhang
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| |
Collapse
|
25
|
Xie ZT, Zeng J, Kang DH, Saito S, Miyagawa S, Sawa Y, Matsusaki M. 3D Printing of Collagen Scaffold with Enhanced Resolution in a Citrate-Modulated Gellan Gum Microgel Bath. Adv Healthc Mater 2023; 12:e2301090. [PMID: 37143444 DOI: 10.1002/adhm.202301090] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/03/2023] [Indexed: 05/06/2023]
Abstract
3D printing in a microgel-based supporting bath enables the construction of complex structures with soft and watery biomaterials but the low print resolution is usually an obstacle to its practical application in tissue engineering. Herein, high-resolution printing of a 3D collagen organ scaffold is realized by using an engineered Gellan gum (GG) microgel bath containing trisodium citrate (TSC). The introduction of TSC into the bath system not only mitigates the aggregation of GG microgels, leading to a more homogeneous bath morphology but also suppresses the diffusion of the collagen ink in the bath due to the dehydration effect of TSC, both of which contribute to the improvement of print resolution. 3D collagen organ structures such as hand, ear, and heart are successfully constructed with high shape fidelity in the developed bath. After printing, the GG and TSC can be easily removed by washing with water, and the obtained collagen product exhibits good cell affinity in a tissue scaffold application. This work offers an easy-to-operate strategy for developing a microgel bath for high-resolution printing of collagen, providing an alternative path to in vitro 3D organ construction.
Collapse
Affiliation(s)
- Zheng-Tian Xie
- Division of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Jinfeng Zeng
- Division of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Dong-Hee Kang
- Division of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeyoshi Saito
- Division of Health Sciences, Department of Medical Physics and Engineering, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Michiya Matsusaki
- Division of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
26
|
Li Y, Zhang X, Zhang X, Zhang Y, Hou D. Recent Progress of the Vat Photopolymerization Technique in Tissue Engineering: A Brief Review of Mechanisms, Methods, Materials, and Applications. Polymers (Basel) 2023; 15:3940. [PMID: 37835989 PMCID: PMC10574968 DOI: 10.3390/polym15193940] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/18/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Vat photopolymerization (VP), including stereolithography (SLA), digital light processing (DLP), and volumetric printing, employs UV or visible light to solidify cell-laden photoactive bioresin contained within a vat in a point-by-point, layer-by-layer, or volumetric manner. VP-based bioprinting has garnered substantial attention in both academia and industry due to its unprecedented control over printing resolution and accuracy, as well as its rapid printing speed. It holds tremendous potential for the fabrication of tissue- and organ-like structures in the field of regenerative medicine. This review summarizes the recent progress of VP in the fields of tissue engineering and regenerative medicine. First, it introduces the mechanism of photopolymerization, followed by an explanation of the printing technique and commonly used biomaterials. Furthermore, the application of VP-based bioprinting in tissue engineering was discussed. Finally, the challenges facing VP-based bioprinting are discussed, and the future trends in VP-based bioprinting are projected.
Collapse
Affiliation(s)
- Ying Li
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Xueqin Zhang
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Xin Zhang
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Yuxuan Zhang
- FuYang Sineva Materials Technology Co., Ltd., Beijing 100176, China
| | - Dan Hou
- Chinese Academy of Meteorological Sciences, China National Petroleum Corporation, Beijing 102206, China
| |
Collapse
|
27
|
Mappa TA, Liu CM, Tseng CC, Ruslin M, Cheng JH, Lan WC, Huang BH, Cho YC, Hsieh CC, Kuo HH, Tsou CH, Shen YK. An Innovative Biofunctional Composite Hydrogel with Enhanced Printability, Rheological Properties, and Structural Integrity for Cell Scaffold Applications. Polymers (Basel) 2023; 15:3223. [PMID: 37571117 PMCID: PMC10421221 DOI: 10.3390/polym15153223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
The present study was conducted to manipulate various biomaterials to find potential hydrogel formulations through three-dimensional (3D) bioprinting fabrication for tissue repair, reconstruction, or regeneration. The hydrogels were prepared using sodium alginate and gelatin combined with different concentrations of Pluronic F127 (6% (3 g), 8% (4 g), and 10% (5 g)) and were marked as AGF-6%, AGF-8%, and AGF-10%, respectively. The properties of the hydrogels were investigated using a contact angle goniometer, rheometer, and 3D bioprinter. In addition, the osteoblast-like cell line (MG-63) was used to evaluate the cell viability including hydrogels before and after 3D bioprinting. It was found that the ratio of contact angle was lowest at AGF-6%, and the rheological results were higher for all samples of AGF-6%, AGF-8%, and AGF-10% compared with the control sample. The printability indicated that the AGF-6% hydrogel possessed great potential in creating a cell scaffold with shape integrity. Moreover, the live/dead assay also presented the highest numbers of live cells before printing compared with after printing. However, the number of live cells on day 7 was higher than on day 1 before and after printing (** p < 0.01). Therefore, the combination of AGF-6% could be developed as a biofunctional hydrogel formulation for potential tissue regeneration applications.
Collapse
Affiliation(s)
- Taufik Abdullah Mappa
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.A.M.)
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Hasanuddin University, Makassar 90245, Indonesia;
| | - Chung-Ming Liu
- Department of Biomedical Engineering, College of Biomedical Engineering, China Medical University, Taichung 404, Taiwan;
| | - Chung-Chih Tseng
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
| | - Muhammad Ruslin
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Hasanuddin University, Makassar 90245, Indonesia;
| | - Jui-Hung Cheng
- Department of Mold and Die Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 807, Taiwan;
| | - Wen-Chien Lan
- Department of Oral Hygiene Care, Ching Kuo Institute of Management and Health, Keelung 203, Taiwan;
| | - Bai-Hung Huang
- Graduate Institute of Dental Science, College of Dentistry, China Medical University, Taichung 404, Taiwan;
| | - Yung-Chieh Cho
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.A.M.)
| | - Chia-Chien Hsieh
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Hsin-Hui Kuo
- Research Center for Biomedical Devices and Prototyping Production, Taipei Medical University, Taipei 110, Taiwan;
| | - Chen-Han Tsou
- Department of Dentistry, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan
| | - Yung-Kang Shen
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan;
| |
Collapse
|
28
|
Wu CA, Zhu Y, Woo YJ. Advances in 3D Bioprinting: Techniques, Applications, and Future Directions for Cardiac Tissue Engineering. Bioengineering (Basel) 2023; 10:842. [PMID: 37508869 PMCID: PMC10376421 DOI: 10.3390/bioengineering10070842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality in the United States. Cardiac tissue engineering is a direction in regenerative medicine that aims to repair various heart defects with the long-term goal of artificially rebuilding a full-scale organ that matches its native structure and function. Three-dimensional (3D) bioprinting offers promising applications through its layer-by-layer biomaterial deposition using different techniques and bio-inks. In this review, we will introduce cardiac tissue engineering, 3D bioprinting processes, bioprinting techniques, bio-ink materials, areas of limitation, and the latest applications of this technology, alongside its future directions for further innovation.
Collapse
Affiliation(s)
- Catherine A Wu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
29
|
Zennifer A, Thangadurai M, Sundaramurthi D, Sethuraman S. Additive manufacturing of peripheral nerve conduits - Fabrication methods, design considerations and clinical challenges. SLAS Technol 2023; 28:102-126. [PMID: 37028493 DOI: 10.1016/j.slast.2023.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
Tissue-engineered nerve guidance conduits (NGCs) are a viable clinical alternative to autografts and allografts and have been widely used to treat peripheral nerve injuries (PNIs). Although these NGCs are successful to some extent, they cannot aid in native regeneration by improving native-equivalent neural innervation or regrowth. Further, NGCs exhibit longer recovery period and high cost limiting their clinical applications. Additive manufacturing (AM) could be an alternative to the existing drawbacks of the conventional NGCs fabrication methods. The emergence of the AM technique has offered ease for developing personalized three-dimensional (3D) neural constructs with intricate features and higher accuracy on a larger scale, replicating the native feature of nerve tissue. This review introduces the structural organization of peripheral nerves, the classification of PNI, and limitations in clinical and conventional nerve scaffold fabrication strategies. The principles and advantages of AM-based techniques, including the combinatorial approaches utilized for manufacturing 3D nerve conduits, are briefly summarized. This review also outlines the crucial parameters, such as the choice of printable biomaterials, 3D microstructural design/model, conductivity, permeability, degradation, mechanical property, and sterilization required to fabricate large-scale additive-manufactured NGCs successfully. Finally, the challenges and future directions toward fabricating the 3D-printed/bioprinted NGCs for clinical translation are also discussed.
Collapse
Affiliation(s)
- Allen Zennifer
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Madhumithra Thangadurai
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| |
Collapse
|
30
|
Afewerki S, Stocco TD, Rosa da Silva AD, Aguiar Furtado AS, Fernandes de Sousa G, Ruiz-Esparza GU, Webster TJ, Marciano FR, Strømme M, Zhang YS, Lobo AO. In vitro high-content tissue models to address precision medicine challenges. Mol Aspects Med 2023; 91:101108. [PMID: 35987701 PMCID: PMC9384546 DOI: 10.1016/j.mam.2022.101108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/29/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2023]
Abstract
The field of precision medicine allows for tailor-made treatments specific to a patient and thereby improve the efficiency and accuracy of disease prevention, diagnosis, and treatment and at the same time would reduce the cost, redundant treatment, and side effects of current treatments. Here, the combination of organ-on-a-chip and bioprinting into engineering high-content in vitro tissue models is envisioned to address some precision medicine challenges. This strategy could be employed to tackle the current coronavirus disease 2019 (COVID-19), which has made a significant impact and paradigm shift in our society. Nevertheless, despite that vaccines against COVID-19 have been successfully developed and vaccination programs are already being deployed worldwide, it will likely require some time before it is available to everyone. Furthermore, there are still some uncertainties and lack of a full understanding of the virus as demonstrated in the high number new mutations arising worldwide and reinfections of already vaccinated individuals. To this end, efficient diagnostic tools and treatments are still urgently needed. In this context, the convergence of bioprinting and organ-on-a-chip technologies, either used alone or in combination, could possibly function as a prominent tool in addressing the current pandemic. This could enable facile advances of important tools, diagnostics, and better physiologically representative in vitro models specific to individuals allowing for faster and more accurate screening of therapeutics evaluating their efficacy and toxicity. This review will cover such technological advances and highlight what is needed for the field to mature for tackling the various needs for current and future pandemics as well as their relevancy towards precision medicine.
Collapse
Affiliation(s)
- Samson Afewerki
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Thiago Domingues Stocco
- Bioengineering Program, Technological and Scientific Institute, Brazil University, 08230-030, São Paulo, SP, Brazil; Faculty of Medical Sciences, Unicamp - State University of Campinas, 13083-877, Campinas, SP, Brazil
| | | | - André Sales Aguiar Furtado
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Gustavo Fernandes de Sousa
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Guillermo U Ruiz-Esparza
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA
| | - Thomas J Webster
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil; Hebei University of Technology, Tianjin, China
| | - Fernanda R Marciano
- Department of Physics, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Maria Strømme
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA.
| | - Anderson Oliveira Lobo
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil.
| |
Collapse
|
31
|
Yang U, Kang B, Yong MJ, Yang DH, Choi SY, Je JH, Oh SS. Type-Independent 3D Writing and Nano-Patterning of Confined Biopolymers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207403. [PMID: 36825681 PMCID: PMC10161081 DOI: 10.1002/advs.202207403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/07/2023] [Indexed: 05/06/2023]
Abstract
Biopolymers are essential building blocks that constitute cells and tissues with well-defined molecular structures and diverse biological functions. Their three-dimensional (3D) complex architectures are used to analyze, control, and mimic various cells and their ensembles. However, the free-form and high-resolution structuring of various biopolymers remain challenging because their structural and rheological control depend critically on their polymeric types at the submicron scale. Here, direct 3D writing of intact biopolymers is demonstrated using a systemic combination of nanoscale confinement, evaporation, and solidification of a biopolymer-containing solution. A femtoliter solution is confined in an ultra-shallow liquid interface between a fine-tuned nanopipette and a chosen substrate surface to achieve directional growth of biopolymer nanowires via solvent-exclusive evaporation and concurrent solution supply. The evaporation-dependent printing is biopolymer type-independent, therefore, the 3D motor-operated precise nanopipette positioning allows in situ printing of nucleic acids, polysaccharides, and proteins with submicron resolution. By controlling concentrations and molecular weights, several different biopolymers are reproducibly patterned with desired size and geometry, and their 3D architectures are biologically active in various solvents with no structural deformation. Notably, protein-based nanowire patterns exhibit pin-point localization of spatiotemporal biofunctions, including target recognition and catalytic peroxidation, indicating their application potential in organ-on-chips and micro-tissue engineering.
Collapse
Affiliation(s)
- Un Yang
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Byunghwa Kang
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Moon-Jung Yong
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Dong-Hwan Yang
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Si-Young Choi
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Jung Ho Je
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
- Nanoblesse, 85-11 (4th fl.) Namwon-Ro, Pohang, Gyeongbuk, 37883, South Korea
| | - Seung Soo Oh
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
- Institute for Convergence Research and Education in Advanced Technology (I-CREATE), Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| |
Collapse
|
32
|
Lam EHY, Yu F, Zhu S, Wang Z. 3D Bioprinting for Next-Generation Personalized Medicine. Int J Mol Sci 2023; 24:ijms24076357. [PMID: 37047328 PMCID: PMC10094501 DOI: 10.3390/ijms24076357] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
In the past decade, immense progress has been made in advancing personalized medicine to effectively address patient-specific disease complexities in order to develop individualized treatment strategies. In particular, the emergence of 3D bioprinting for in vitro models of tissue and organ engineering presents novel opportunities to improve personalized medicine. However, the existing bioprinted constructs are not yet able to fulfill the ultimate goal: an anatomically realistic organ with mature biological functions. Current bioprinting approaches have technical challenges in terms of precise cell deposition, effective differentiation, proper vascularization, and innervation. This review introduces the principles and realizations of bioprinting with a strong focus on the predominant techniques, including extrusion printing and digital light processing (DLP). We further discussed the applications of bioprinted constructs, including the engraftment of stem cells as personalized implants for regenerative medicine and in vitro high-throughput drug development models for drug discovery. While no one-size-fits-all approach to bioprinting has emerged, the rapid progress and promising results of preliminary studies have demonstrated that bioprinting could serve as an empowering technology to resolve critical challenges in personalized medicine.
Collapse
Affiliation(s)
- Ethan Hau Yin Lam
- Faculty of Arts and Science, University of Toronto, Toronto, ON M5S 3G3, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Fengqing Yu
- Faculty of Arts and Science, University of Toronto, Toronto, ON M5S 3G3, Canada
- Department of Computer Science, University of Toronto, Toronto, ON M5S 1A4, Canada
| | - Sabrina Zhu
- Faculty of Arts and Science, University of Toronto, Toronto, ON M5S 3G3, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Zongjie Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1, Canada
- McCormick School of Engineering, Northwestern University, Chicago, IL 60611, USA
- Correspondence: or
| |
Collapse
|
33
|
Application of 3D Printing in Bone Grafts. Cells 2023; 12:cells12060859. [PMID: 36980200 PMCID: PMC10047278 DOI: 10.3390/cells12060859] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
The application of 3D printing in bone grafts is gaining in importance and is becoming more and more popular. The choice of the method has a direct impact on the preparation of the patient for surgery, the probability of rejection of the transplant, and many other complications. The aim of the article is to discuss methods of bone grafting and to compare these methods. This review of literature is based on a selective literature search of the PubMed and Web of Science databases from 2001 to 2022 using the search terms “bone graft”, “bone transplant”, and “3D printing”. In addition, we also reviewed non-medical literature related to materials used for 3D printing. There are several methods of bone grafting, such as a demineralized bone matrix, cancellous allograft, nonvascular cortical allograft, osteoarticular allograft, osteochondral allograft, vascularized allograft, and an autogenic transplant using a bone substitute. Currently, autogenous grafting, which involves removing the patient’s bone from an area of low aesthetic importance, is referred to as the gold standard. 3D printing enables using a variety of materials. 3D technology is being applied to bone tissue engineering much more often. It allows for the treatment of bone defects thanks to the creation of a porous scaffold with adequate mechanical strength and favorable macro- and microstructures. Bone tissue engineering is an innovative approach that can be used to repair multiple bone defects in the process of transplantation. In this process, biomaterials are a very important factor in supporting regenerative cells and the regeneration of tissue. We have years of research ahead of us; however, it is certain that 3D printing is the future of transplant medicine.
Collapse
|
34
|
Mori S, Ito T, Takao H, Shimokawa F, Terao K. Optically driven microtools with an antibody-immobilised surface for on-site cell assembly. IET Nanobiotechnol 2023; 17:197-203. [PMID: 36647211 DOI: 10.1049/nbt2.12114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/21/2022] [Accepted: 12/27/2022] [Indexed: 01/18/2023] Open
Abstract
To enable the accurate reproduction of organs in vitro, and improve drug screening efficiency and regenerative medicine research, it is necessary to assemble cells with single-cell resolution to form cell clusters. However, a method to assemble such forms has not been developed. In this study, a platform for on-site cell assembly at the single-cell level using optically driven microtools in a microfluidic device is developed. The microtool was fabricated by SU-8 photolithography, and antibodies were immobilised on its surface. The cells were captured by the microtool through the bindings between the antibodies on the microtool and the antigens on the cell membrane. Transmembrane proteins, CD51/61 and CD44 that facilitate cell adhesion, commonly found on the surface of cancer cells were targeted. The microtool containing antibodies for CD51/61 and CD44 proteins was manipulated using optical tweezers to capture HeLa cells placed on a microfluidic device. A comparison of the adhesion rates of different surface treatments showed the superiority of the antibody-immobilised microtool. The assembly of multiple cells into a cluster by repeating the cell capture process is further demonstrated. The geometry and surface function of the microtool can be modified according to the cell assembly requirements. The platform can be used in regenerative medicine and drug screening to produce cell clusters that closely resemble tissues and organs in vivo.
Collapse
Affiliation(s)
- Shuntaro Mori
- Department of Intelligent Mechanical Systems Engineering, Kagawa University, Takamatsu, Japan
| | - Takumi Ito
- Department of Intelligent Mechanical Systems Engineering, Kagawa University, Takamatsu, Japan
| | - Hidekuni Takao
- Department of Intelligent Mechanical Systems Engineering, Kagawa University, Takamatsu, Japan.,Nano-Micro Structure Device Integrated Research Center, Kagawa University, Takamatsu, Japan
| | - Fusao Shimokawa
- Department of Intelligent Mechanical Systems Engineering, Kagawa University, Takamatsu, Japan.,Nano-Micro Structure Device Integrated Research Center, Kagawa University, Takamatsu, Japan
| | - Kyohei Terao
- Department of Intelligent Mechanical Systems Engineering, Kagawa University, Takamatsu, Japan.,Nano-Micro Structure Device Integrated Research Center, Kagawa University, Takamatsu, Japan
| |
Collapse
|
35
|
Assad H, Assad A, Kumar A. Recent Developments in 3D Bio-Printing and Its Biomedical Applications. Pharmaceutics 2023; 15:255. [PMID: 36678884 PMCID: PMC9861443 DOI: 10.3390/pharmaceutics15010255] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
The fast-developing field of 3D bio-printing has been extensively used to improve the usability and performance of scaffolds filled with cells. Over the last few decades, a variety of tissues and organs including skin, blood vessels, and hearts, etc., have all been produced in large quantities via 3D bio-printing. These tissues and organs are not only able to serve as building blocks for the ultimate goal of repair and regeneration, but they can also be utilized as in vitro models for pharmacokinetics, drug screening, and other purposes. To further 3D-printing uses in tissue engineering, research on novel, suitable biomaterials with quick cross-linking capabilities is a prerequisite. A wider variety of acceptable 3D-printed materials are still needed, as well as better printing resolution (particularly at the nanoscale range), speed, and biomaterial compatibility. The aim of this study is to provide expertise in the most prevalent and new biomaterials used in 3D bio-printing as well as an introduction to the associated approaches that are frequently considered by researchers. Furthermore, an effort has been made to convey the most pertinent implementations of 3D bio-printing processes, such as tissue regeneration, etc., by providing the most significant research together with a comprehensive list of material selection guidelines, constraints, and future prospects.
Collapse
Affiliation(s)
- Humira Assad
- Department of Chemistry, School of Chemical Engineering and Physical Sciences, Lovely Professional University, Punjab 144001, India
| | - Arvina Assad
- Bibi Halima College of Nursing and Medical Technology, Srinagar 190010, India
| | - Ashish Kumar
- Nalanda College of Engineering, Department of Science and Technology, Government of Bihar, Patna 803108, India
| |
Collapse
|
36
|
Hrynevich A, Li Y, Cedillo-Servin G, Malda J, Castilho M. (Bio)fabrication of microfluidic devices and organs-on-a-chip. 3D Print Med 2023. [DOI: 10.1016/b978-0-323-89831-7.00001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
37
|
Augustine R, Gezek M, Seray Bostanci N, Nguyen A, Camci-Unal G. Oxygen-Generating Scaffolds: One Step Closer to the Clinical Translation of Tissue Engineered Products. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2023; 455:140783. [PMID: 36644784 PMCID: PMC9835968 DOI: 10.1016/j.cej.2022.140783] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The lack of oxygen supply in engineered constructs has been an ongoing challenge for tissue engineering and regenerative medicine. Upon implantation of an engineered tissue, spontaneous blood vessel formation does not happen rapidly, therefore, there is typically a limited availability of oxygen in engineered biomaterials. Providing oxygen in large tissue-engineered constructs is a major challenge that hinders the development of clinically relevant engineered tissues. Similarly, maintaining adequate oxygen levels in cell-laden tissue engineered products during transportation and storage is another hurdle. There is an unmet demand for functional scaffolds that could actively produce and deliver oxygen, attainable by incorporating oxygen-generating materials. Recent approaches include encapsulation of oxygen-generating agents such as solid peroxides, liquid peroxides, and fluorinated substances in the scaffolds. Recent approaches to mitigate the adverse effects, as well as achieving a sustained and controlled release of oxygen, are discussed. Importance of oxygen-generating materials in various tissue engineering approaches such as ex vivo tissue engineering, in situ tissue engineering, and bioprinting are highlighted in detail. In addition, the existing challenges, possible solutions, and future strategies that aim to design clinically relevant multifunctional oxygen-generating biomaterials are provided in this review paper.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Mert Gezek
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Nazli Seray Bostanci
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Angelina Nguyen
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
38
|
Bolívar-Monsalve EJ, Ceballos-González CF, Chávez-Madero C, de la Cruz-Rivas BG, Velásquez Marín S, Mora-Godínez S, Reyes-Cortés LM, Khademhosseini A, Weiss PS, Samandari M, Tamayol A, Alvarez MM, Trujillo-de Santiago G. One-Step Bioprinting of Multi-Channel Hydrogel Filaments Using Chaotic Advection: Fabrication of Pre-Vascularized Muscle-Like Tissues. Adv Healthc Mater 2022; 11:e2200448. [PMID: 35930168 DOI: 10.1002/adhm.202200448] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/07/2022] [Indexed: 01/28/2023]
Abstract
The biofabrication of living constructs containing hollow channels is critical for manufacturing thick tissues. However, current technologies are limited in their effectiveness in the fabrication of channels with diameters smaller than hundreds of micrometers. It is demonstrated that the co-extrusion of cell-laden hydrogels and sacrificial materials through printheads containing Kenics static mixing elements enables the continuous and one-step fabrication of thin hydrogel filaments (1 mm in diameter) containing dozens of hollow microchannels with widths as small as a single cell. Pre-vascularized skeletal muscle-like filaments are bioprinted by loading murine myoblasts (C2C12 cells) in gelatin methacryloyl - alginate hydrogels and using hydroxyethyl cellulose as a sacrificial material. Higher viability and metabolic activity are observed in filaments with hollow multi-channels than in solid constructs. The presence of hollow channels promotes the expression of Ki67 (a proliferation biomarker), mitigates the expression of hypoxia-inducible factor 1-alpha , and markedly enhances cell alignment (i.e., 82% of muscle myofibrils aligned (in ±10°) to the main direction of the microchannels after seven days of culture). The emergence of sarcomeric α-actin is verified through immunofluorescence and gene expression. Overall, this work presents an effective and practical tool for the fabrication of pre-vascularized engineered tissues.
Collapse
Affiliation(s)
| | | | - Carolina Chávez-Madero
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | - Brenda Guadalupe de la Cruz-Rivas
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | - Silvana Velásquez Marín
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | - Shirley Mora-Godínez
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| | - Paul S Weiss
- Department of Chemistry and Biochemistry, Department of Bioengineering, Department of Materials Science and Engineering, California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Mario Moisés Alvarez
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | - Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| |
Collapse
|
39
|
Emerging biomaterials and technologies to control stem cell fate and patterning in engineered 3D tissues and organoids. Biointerphases 2022; 17:060801. [DOI: 10.1116/6.0002034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The ability to create complex three-dimensional cellular models that can effectively replicate the structure and function of human organs and tissues in vitro has the potential to revolutionize medicine. Such models could facilitate the interrogation of developmental and disease processes underpinning fundamental discovery science, vastly accelerate drug development and screening, or even be used to create tissues for implantation into the body. Realization of this potential, however, requires the recreation of complex biochemical, biophysical, and cellular patterns of 3D tissues and remains a key challenge in the field. Recent advances are being driven by improved knowledge of tissue morphogenesis and architecture and technological developments in bioengineering and materials science that can create the multidimensional and dynamic systems required to produce complex tissue microenvironments. In this article, we discuss challenges for in vitro models of tissues and organs and summarize the current state-of-the art in biomaterials and bioengineered systems that aim to address these challenges. This includes both top-down technologies, such as 3D photopatterning, magnetism, acoustic forces, and cell origami, as well as bottom-up patterning using 3D bioprinting, microfluidics, cell sheet technology, or composite scaffolds. We illustrate the varying ways that these can be applied to suit the needs of different tissues and applications by focussing on specific examples of patterning the bone-tendon interface, kidney organoids, and brain cancer models. Finally, we discuss the challenges and future prospects in applying materials science and bioengineering to develop high-quality 3D tissue structures for in vitro studies.
Collapse
|
40
|
Zhai H, Jin X, Minnick G, Rosenbohm J, Hafiz MAH, Yang R, Meng F. Spatially Guided Construction of Multilayered Epidermal Models Recapturing Structural Hierarchy and Cell-Cell Junctions. SMALL SCIENCE 2022; 2:2200051. [PMID: 36590765 PMCID: PMC9799093 DOI: 10.1002/smsc.202200051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
A current challenge in three-dimensional (3D) bioprinting of skin equivalents is to recreate the distinct basal and suprabasal layers and to promote their direct interactions. Such a structural arrangement is essential to establish 3D stratified epidermis disease models, such as for the autoimmune skin disease pemphigus vulgaris (PV), which targets the cell-cell junctions at the interface of the basal and suprabasal layers. Inspired by epithelial regeneration in wound healing, we develop a method that combines 3D bioprinting and spatially guided self-reorganization of keratinocytes to recapture the fine structural hierarchy that lies in the deep layers of the epidermis. Here, keratinocyte-laden fibrin hydrogels are bioprinted to create geographical cues, guiding dynamic self-reorganization of cells through collective migration, keratinocyte differentiation and vertical expansion. This process results in a region of self-organized multilayers (SOMs) that contain the basal to suprabasal transition, marked by the expressed levels of different types of keratins that indicate differentiation. Finally, we demonstrate the reconstructed skin tissue as an in vitro platform to study the pathogenic effects of PV and observe a significant difference in cell-cell junction dissociation from PV antibodies in different epidermis layers, indicating their applications in the preclinical test of possible therapies.
Collapse
Affiliation(s)
- Haiwei Zhai
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Grayson Minnick
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | | | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Fanben Meng
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
41
|
Sufaru IG, Macovei G, Stoleriu S, Martu MA, Luchian I, Kappenberg-Nitescu DC, Solomon SM. 3D Printed and Bioprinted Membranes and Scaffolds for the Periodontal Tissue Regeneration: A Narrative Review. MEMBRANES 2022; 12:membranes12090902. [PMID: 36135920 PMCID: PMC9505571 DOI: 10.3390/membranes12090902] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 05/31/2023]
Abstract
Numerous technologies and materials were developed with the aim of repairing and reconstructing the tissue loss in patients with periodontitis. Periodontal guided bone regeneration (GBR) and guided tissue regeneration (GTR) involves the use of a membrane which prevents epithelial cell migration, and helps to maintain the space, creating a protected area in which tissue regeneration is favored. Over the time, manufacturing procedures of such barrier membranes followed important improvements. Three-dimensional (3D) printing technology has led to major innovations in periodontal regeneration methods, using technologies such as inkjet printing, light-assisted 3D printing or micro-extrusion. Besides the 3D printing of monophasic and multi-phasic scaffolds, bioprinting and tissue engineering have emerged as innovative technologies which can change the way we see GTR and GBR.
Collapse
Affiliation(s)
- Irina-Georgeta Sufaru
- Department of Periodontology, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| | - Georgiana Macovei
- Department of Oral and Dental Diagnostics, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| | - Simona Stoleriu
- Department of Cariology and Restorative Dental Therapy, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| | - Maria-Alexandra Martu
- Department of Periodontology, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| | - Ionut Luchian
- Department of Periodontology, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| | | | - Sorina Mihaela Solomon
- Department of Periodontology, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| |
Collapse
|
42
|
Chen C, Wang Y, Zhang H, Zhang H, Dong W, Sun W, Zhao Y. Responsive and self-healing structural color supramolecular hydrogel patch for diabetic wound treatment. Bioact Mater 2022; 15:194-202. [PMID: 35386338 PMCID: PMC8940762 DOI: 10.1016/j.bioactmat.2021.11.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/11/2021] [Accepted: 11/27/2021] [Indexed: 12/15/2022] Open
Abstract
The treatment of diabetic wounds remains a great challenge for medical community. Here, we present a novel structural color supramolecular hydrogel patch for diabetic wound treatment. This hydrogel patch was created by using N-acryloyl glycinamide (NAGA) and 1-vinyl-1,2,4-triazole (VTZ) mixed supramolecular hydrogel as the inverse opal scaffold, and temperature responsive poly(N-isopropylacrylamide) (PNIPAM) hydrogel loaded with vascular endothelial cell growth factor (VEGF) as a filler. Supramolecular hydrogel renders hydrogel patch with superior mechanical properties, in which NAGA and VTZ also provide self-healing and antibacterial properties, respectively. Besides, as the existence of PNIPAM, the hydrogel patch was endowed with thermal-responsiveness property, which could release actives in response to temperature stimulus. Given these excellent performances, we have demonstrated that the supramolecular hydrogel patch could significantly enhance the wound healing process in diabetes rats by downregulating the expression of inflammatory factors, promoting collagen deposition and angiogenesis. Attractively, due to responsive optical property of inverse opal scaffold, the hydrogel patch could display color-sensing behavior that was suitable for the wound monitoring and management as well as guidance of clinical treatment. These distinctive features indicate that the presented hydrogel patches have huge potential values in biomedical fields.
Collapse
Affiliation(s)
- Canwen Chen
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yu Wang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Han Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Hui Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Weiliang Dong
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211800, China
| | - Weijian Sun
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
43
|
Chen Y, Zhang C, Zhang S, Qi H, Zhang D, Li Y, Fang J. Novel advances in strategies and applications of artificial articular cartilage. Front Bioeng Biotechnol 2022; 10:987999. [PMID: 36072291 PMCID: PMC9441570 DOI: 10.3389/fbioe.2022.987999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022] Open
Abstract
Artificial articular cartilage (AC) is extensively applied in the repair and regeneration of cartilage which lacks self-regeneration capacity because of its avascular and low-cellularity nature. With advances in tissue engineering, bioengineering techniques for artificial AC construction have been increasing and maturing gradually. In this review, we elaborated on the advances of biological scaffold technologies in artificial AC including freeze-drying, electrospinning, 3D bioprinting and decellularized, and scaffold-free methods such as self-assembly and cell sheet. In the following, several successful applications of artificial AC built by scaffold and scaffold-free techniques are introduced to demonstrate the clinical application value of artificial AC.
Collapse
Affiliation(s)
- Yifei Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenyue Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shiyong Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hexu Qi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jie Fang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie Fang,
| |
Collapse
|
44
|
A Three-Dimensional Bioprinted Copolymer Scaffold with Biocompatibility and Structural Integrity for Potential Tissue Regeneration Applications. Polymers (Basel) 2022; 14:polym14163415. [PMID: 36015671 PMCID: PMC9413511 DOI: 10.3390/polym14163415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
The present study was to investigate the rheological property, printability, and cell viability of alginate−gelatin composed hydrogels as a potential cell-laden bioink for three-dimensional (3D) bioprinting applications. The 2 g of sodium alginate dissolved in 50 mL of phosphate buffered saline solution was mixed with different concentrations (1% (0.5 g), 2% (1 g), 3% (1.5 g), and 4% (2 g)) of gelatin, denoted as GBH-1, GBH-2, GBH-3, and GBH-4, respectively. The properties of the investigated hydrogels were characterized by contact angle goniometer, rheometer, and bioprinter. In addition, the hydrogel with a proper concentration was adopted as a cell-laden bioink to conduct cell viability testing (before and after bioprinting) using Live/Dead assay and immunofluorescence staining with a human corneal fibroblast cell line. The analytical results indicated that the GBH-2 hydrogel exhibited the lowest loss rate of contact angle (28%) and similar rheological performance as compared with other investigated hydrogels and the control group. Printability results also showed that the average wire diameter of the GBH-2 bioink (0.84 ± 0.02 mm (*** p < 0.001)) post-printing was similar to that of the control group (0.79 ± 0.05 mm). Moreover, a cell scaffold could be fabricated from the GBH-2 bioink and retained its shape integrity for 24 h post-printing. For bioprinting evaluation, it demonstrated that the GBH-2 bioink possessed well viability (>70%) of the human corneal fibroblast cell after seven days of printing under an ideal printing parameter combination (0.4 mm of inner diameter needle, 0.8 bar of printing pressure, and 25 °C of printing temperature). Therefore, the present study suggests that the GBH-2 hydrogel could be developed as a potential cell-laden bioink to print a cell scaffold with biocompatibility and structural integrity for soft tissues such as skin, cornea, nerve, and blood vessel regeneration applications.
Collapse
|
45
|
Bucciarelli A, Motta A. Use of Bombyx mori silk fibroin in tissue engineering: From cocoons to medical devices, challenges, and future perspectives. BIOMATERIALS ADVANCES 2022; 139:212982. [PMID: 35882138 DOI: 10.1016/j.bioadv.2022.212982] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 05/26/2023]
Abstract
Silk fibroin has become a prominent material in tissue engineering (TE) over the last 20 years with almost 10,000 published works spanning in all the TE applications, from skeleton to neuronal regeneration. Fibroin is an extremely versatile biopolymer that, due to its ease of processing, has enabled the development of an entire plethora of materials whose properties and architectures can be tailored to suit target applications. Although the research and development of fibroin TE materials and devices is mature, apart from sutures, only a few medical products made of fibroin are used in the clinical routines. <40 clinical trials of Bombyx mori silk-related products have been reported by the FDA and few of them resulted in a commercialized device. In this review, after explaining the structure and properties of silk fibroin, we provide an overview of both fibroin constructs existing in the literature and fibroin devices used in clinic. Through the comparison of these two categories, we identified the burning issues faced by fibroin products during their translation to the market. Two main aspects will be considered. The first is the standardization of production processes, which leads both to the standardization of the characteristics of the issued device and the correct assessment of its failure. The second is the FDA regulations, which allow new devices to be marketed through the 510(k) clearance by demonstrating their equivalence to a commercialized medical product. The history of some fibroin medical devices will be taken as a case study. Finally, we will outline a roadmap outlining what actions we believe are needed to bring fibroin products to the market.
Collapse
Affiliation(s)
- Alessio Bucciarelli
- CNR nanotech, National Council of Research, University Campus Ecotekne, Via Monteroni, 73100 Lecce, Italy.
| | - Antonella Motta
- BIOtech research centre and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Department of Industrial Engineering, University of Trento, Via delle Regole 101, 38123 Trento, Italy.
| |
Collapse
|
46
|
Jain P, Kathuria H, Dubey N. Advances in 3D bioprinting of tissues/organs for regenerative medicine and in-vitro models. Biomaterials 2022; 287:121639. [PMID: 35779481 DOI: 10.1016/j.biomaterials.2022.121639] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/05/2022] [Accepted: 06/14/2022] [Indexed: 11/24/2022]
Abstract
Tissue/organ shortage is a major medical challenge due to donor scarcity and patient immune rejections. Furthermore, it is difficult to predict or mimic the human disease condition in animal models during preclinical studies because disease phenotype differs between humans and animals. Three-dimensional bioprinting (3DBP) is evolving into an unparalleled multidisciplinary technology for engineering three-dimensional (3D) biological tissue with complex architecture and composition. The technology has emerged as a key driver by precise deposition and assembly of biomaterials with patient's/donor cells. This advancement has aided in the successful fabrication of in vitro models, preclinical implants, and tissue/organs-like structures. Here, we critically reviewed the current state of 3D-bioprinting strategies for regenerative therapy in eight organ systems, including nervous, cardiovascular, skeletal, integumentary, endocrine and exocrine, gastrointestinal, respiratory, and urinary systems. We also focus on the application of 3D bioprinting to fabricated in vitro models to study cancer, infection, drug testing, and safety assessment. The concept of in situ 3D bioprinting is discussed, which is the direct printing of tissues at the injury or defect site for reparative and regenerative therapy. Finally, issues such as scalability, immune response, and regulatory approval are discussed, as well as recently developed tools and technologies such as four-dimensional and convergence bioprinting. In addition, information about clinical trials using 3D printing has been included.
Collapse
Affiliation(s)
- Pooja Jain
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, Maharashtra, India; Faculty of Dentistry, National University of Singapore, Singapore
| | - Himanshu Kathuria
- Department of Pharmacy, National University of Singapore, 117543, Singapore; Nusmetic Pte Ltd, Makerspace, I4 Building, 3 Research Link Singapore, 117602, Singapore.
| | - Nileshkumar Dubey
- Faculty of Dentistry, National University of Singapore, Singapore; ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore.
| |
Collapse
|
47
|
Nieto D, Jiménez G, Moroni L, López-Ruiz E, Gálvez-Martín P, Marchal JA. Biofabrication approaches and regulatory framework of metastatic tumor-on-a-chip models for precision oncology. Med Res Rev 2022; 42:1978-2001. [PMID: 35707911 PMCID: PMC9545141 DOI: 10.1002/med.21914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 12/14/2022]
Abstract
The complexity of the tumor microenvironment (TME) together with the development of the metastatic process are the main reasons for the failure of conventional anticancer treatment. In recent years, there is an increasing need to advance toward advanced in vitro models of cancer mimicking TME and simulating metastasis to understand the associated mechanisms that are still unknown, and to be able to develop personalized therapy. In this review, the commonly used alternatives and latest advances in biofabrication of tumor‐on‐chips, which allow the generation of the most sophisticated and optimized models for recapitulating the tumor process, are presented. In addition, the advances that have allowed these new models in the area of metastasis, cancer stem cells, and angiogenesis are summarized, as well as the recent integration of multiorgan‐on‐a‐chip systems to recapitulate natural metastasis and pharmacological screening against it. We also analyze, for the first time in the literature, the normative and regulatory framework in which these models could potentially be found, as well as the requirements and processes that must be fulfilled to be commercially implemented as in vitro study model. Moreover, we are focused on the possible regulatory pathways for their clinical application in precision medicine and decision making through the generation of personalized models with patient samples. In conclusion, this review highlights the synergistic combination of three‐dimensional bioprinting systems with the novel tumor/metastasis/multiorgan‐on‐a‐chip systems to generate models for both basic research and clinical applications to have devices useful for personalized oncology.
Collapse
Affiliation(s)
- Daniel Nieto
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, University of Maastricht, Universiteitssingel, Maastricht, The Netherlands.,Center for Biomedical Research (CIBM)/Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain
| | - Gema Jiménez
- Center for Biomedical Research (CIBM)/Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, University of Maastricht, Universiteitssingel, Maastricht, The Netherlands
| | - Elena López-Ruiz
- Center for Biomedical Research (CIBM)/Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain.,Department of Health Sciences, University of Jaén, Jaén, Spain
| | | | - Juan Antonio Marchal
- Center for Biomedical Research (CIBM)/Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| |
Collapse
|
48
|
Osouli-Bostanabad K, Masalehdan T, Kapsa RMI, Quigley A, Lalatsa A, Bruggeman KF, Franks SJ, Williams RJ, Nisbet DR. Traction of 3D and 4D Printing in the Healthcare Industry: From Drug Delivery and Analysis to Regenerative Medicine. ACS Biomater Sci Eng 2022; 8:2764-2797. [PMID: 35696306 DOI: 10.1021/acsbiomaterials.2c00094] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Three-dimensional (3D) printing and 3D bioprinting are promising technologies for a broad range of healthcare applications from frontier regenerative medicine and tissue engineering therapies to pharmaceutical advancements yet must overcome the challenges of biocompatibility and resolution. Through comparison of traditional biofabrication methods with 3D (bio)printing, this review highlights the promise of 3D printing for the production of on-demand, personalized, and complex products that enhance the accessibility, effectiveness, and safety of drug therapies and delivery systems. In addition, this review describes the capacity of 3D bioprinting to fabricate patient-specific tissues and living cell systems (e.g., vascular networks, organs, muscles, and skeletal systems) as well as its applications in the delivery of cells and genes, microfluidics, and organ-on-chip constructs. This review summarizes how tailoring selected parameters (i.e., accurately selecting the appropriate printing method, materials, and printing parameters based on the desired application and behavior) can better facilitate the development of optimized 3D-printed products and how dynamic 4D-printed strategies (printing materials designed to change with time or stimulus) may be deployed to overcome many of the inherent limitations of conventional 3D-printed technologies. Comprehensive insights into a critical perspective of the future of 4D bioprinting, crucial requirements for 4D printing including the programmability of a material, multimaterial printing methods, and precise designs for meticulous transformations or even clinical applications are also given.
Collapse
Affiliation(s)
- Karim Osouli-Bostanabad
- Biomaterials, Bio-engineering and Nanomedicine (BioN) Lab, Institute of Biomedical and Biomolecular, Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Tahereh Masalehdan
- Department of Materials Engineering, Institute of Mechanical Engineering, University of Tabriz, Tabriz 51666-16444, Iran
| | - Robert M I Kapsa
- Biomedical and Electrical Engineering, School of Engineering, RMIT University, Melbourne, Victoria 3000, Australia.,Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Anita Quigley
- Biomedical and Electrical Engineering, School of Engineering, RMIT University, Melbourne, Victoria 3000, Australia.,Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Aikaterini Lalatsa
- Biomaterials, Bio-engineering and Nanomedicine (BioN) Lab, Institute of Biomedical and Biomolecular, Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Kiara F Bruggeman
- Laboratory of Advanced Biomaterials, Research School of Chemistry and the John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory 2601, Australia.,Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Stephanie J Franks
- Laboratory of Advanced Biomaterials, Research School of Chemistry and the John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Richard J Williams
- Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - David R Nisbet
- Laboratory of Advanced Biomaterials, Research School of Chemistry and the John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory 2601, Australia.,The Graeme Clark Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia.,Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
49
|
Hu M, Ling Z, Ren X. Extracellular matrix dynamics: tracking in biological systems and their implications. J Biol Eng 2022; 16:13. [PMID: 35637526 PMCID: PMC9153193 DOI: 10.1186/s13036-022-00292-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/11/2022] [Indexed: 12/23/2022] Open
Abstract
The extracellular matrix (ECM) constitutes the main acellular microenvironment of cells in almost all tissues and organs. The ECM not only provides mechanical support, but also mediates numerous biochemical interactions to guide cell survival, proliferation, differentiation, and migration. Thus, better understanding the everchanging temporal and spatial shifts in ECM composition and structure - the ECM dynamics - will provide fundamental insight regarding extracellular regulation of tissue homeostasis and how tissue states transition from one to another during diverse pathophysiological processes. This review outlines the mechanisms mediating ECM-cell interactions and highlights how changes in the ECM modulate tissue development and disease progression, using the lung as the primary model organ. We then discuss existing methodologies for revealing ECM compositional dynamics, with a particular focus on tracking newly synthesized ECM proteins. Finally, we discuss the ramifications ECM dynamics have on tissue engineering and how to implement spatial and temporal specific extracellular microenvironments into bioengineered tissues. Overall, this review communicates the current capabilities for studying native ECM dynamics and delineates new research directions in discovering and implementing ECM dynamics to push the frontier forward.
Collapse
Affiliation(s)
- Michael Hu
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA, 15213, USA
| | - Zihan Ling
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA, 15213, USA
| | - Xi Ren
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
50
|
Liu S, Wang T, Li S, Wang X. Application Status of Sacrificial Biomaterials in 3D Bioprinting. Polymers (Basel) 2022; 14:2182. [PMID: 35683853 PMCID: PMC9182955 DOI: 10.3390/polym14112182] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
Additive manufacturing, also known as three-dimensional (3D) printing, relates to several rapid prototyping (RP) technologies, and has shown great potential in the manufacture of organoids and even complex bioartificial organs. A major challenge for 3D bioprinting complex org unit ans is the competitive requirements with respect to structural biomimeticability, material integrability, and functional manufacturability. Over the past several years, 3D bioprinting based on sacrificial templates has shown its unique advantages in building hierarchical vascular networks in complex organs. Sacrificial biomaterials as supporting structures have been used widely in the construction of tubular tissues. The advent of suspension printing has enabled the precise printing of some soft biomaterials (e.g., collagen and fibrinogen), which were previously considered unprintable singly with cells. In addition, the introduction of sacrificial biomaterials can improve the porosity of biomaterials, making the printed structures more favorable for cell proliferation, migration and connection. In this review, we mainly consider the latest developments and applications of 3D bioprinting based on the strategy of sacrificial biomaterials, discuss the basic principles of sacrificial templates, and look forward to the broad prospects of this approach for complex organ engineering or manufacturing.
Collapse
Affiliation(s)
- Siyu Liu
- Center of 3D Printing & Organ Manufacturing, School of Intelligent Medicine, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China; (S.L.); (T.W.); (S.L.)
| | - Tianlin Wang
- Center of 3D Printing & Organ Manufacturing, School of Intelligent Medicine, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China; (S.L.); (T.W.); (S.L.)
| | - Shenglong Li
- Center of 3D Printing & Organ Manufacturing, School of Intelligent Medicine, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China; (S.L.); (T.W.); (S.L.)
| | - Xiaohong Wang
- Center of 3D Printing & Organ Manufacturing, School of Intelligent Medicine, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China; (S.L.); (T.W.); (S.L.)
- Center of Organ Manufacturing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|