1
|
Jiang Z, Jin B, Liang Z, Wang Y, Ren S, Huang Y, Li C, Sun H, Li Y, Liu L, Li N, Wang J, Cui Z, Huang P, Yang H, Mao Y, Ye H. Liver bioprinting within a novel support medium with functionalized spheroids, hepatic vein structures, and enhanced post-transplantation vascularization. Biomaterials 2024; 311:122681. [PMID: 38944968 DOI: 10.1016/j.biomaterials.2024.122681] [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: 12/19/2023] [Revised: 04/28/2024] [Accepted: 06/23/2024] [Indexed: 07/02/2024]
Abstract
Cell-laden bioprinting is a promising biofabrication strategy for regenerating bioactive transplants to address organ donor shortages. However, there has been little success in reproducing transplantable artificial organs with multiple distinctive cell types and physiologically relevant architecture. In this study, an omnidirectional printing embedded network (OPEN) is presented as a support medium for embedded 3D printing. The medium is state-of-the-art due to its one-step preparation, fast removal, and versatile ink compatibility. To test the feasibility of OPEN, exceptional primary mouse hepatocytes (PMHs) and endothelial cell line-C166, were used to print hepatospheroid-encapsulated-artificial livers (HEALs) with vein structures following predesigned anatomy-based printing paths in OPEN. PMHs self-organized into hepatocyte spheroids within the ink matrix, whereas the entire cross-linked structure remained intact for a minimum of ten days of cultivation. Cultivated HEALs maintained mature hepatic functions and marker gene expression at a higher level than conventional 2D and 3D conditions in vitro. HEALs with C166-laden vein structures promoted endogenous neovascularization in vivo compared with hepatospheroid-only liver prints within two weeks of transplantation. Collectively, the proposed platform enables the manufacture of bioactive tissues or organs resembling anatomical architecture, and has broad implications for liver function replacement in clinical applications.
Collapse
Affiliation(s)
- Zhuoran Jiang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China; Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Zhu Liang
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK; Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Yinhan Wang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Shuai Ren
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Yongfa Huang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Changcan Li
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Hang Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Yunzhu Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Li Liu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Nianlin Li
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Jinzhuo Wang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK; The Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou, 215123, China
| | - Pengyu Huang
- Engineering Research Center of Pulmonary and Critical Care Technology and Device (MOE of China), Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China.
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China.
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China.
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK; The Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou, 215123, China.
| |
Collapse
|
2
|
Beilharz S, Debnath MK, Vinella D, Shoffstall AJ, Karayilan M. Advances in Injectable Polymeric Biomaterials and Their Contemporary Medical Practices. ACS APPLIED BIO MATERIALS 2024. [PMID: 39471414 DOI: 10.1021/acsabm.4c01001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Injectable biomaterials have been engineered to operate within the human body, offering versatile solutions for minimally invasive therapies and meeting several stringent requirements such as biocompatibility, biodegradability, low viscosity for ease of injection, mechanical strength, rapid gelation postinjection, controlled release of therapeutic agents, hydrophobicity/hydrophilicity balance, stability under physiological conditions, and the ability to be sterilized. Their adaptability and performance in diverse clinical settings make them invaluable for modern medical treatments. This article reviews recent advancements in the design, synthesis, and characterization of injectable polymeric biomaterials, providing insights into their emerging applications. We discuss a broad spectrum of these materials, including natural, synthetic, hybrid, and composite types, that are being applied in targeted drug delivery, cell and protein transport, regenerative medicine, tissue adhesives, injectable implants, bioimaging, diagnostics, and 3D bioprinting. Ultimately, the review highlights the critical role of injectable polymeric biomaterials in shaping the future of medical treatments and improving patient outcomes across a wide range of therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Sophia Beilharz
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Mithun Kumar Debnath
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Daniele Vinella
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Metin Karayilan
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
3
|
de Souza Araújo IJ, Perkins RS, Ibrahim MM, Huang GTJ, Zhang W. Bioprinting PDLSC-Laden Collagen Scaffolds for Periodontal Ligament Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39467547 DOI: 10.1021/acsami.4c13830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Periodontitis and severe trauma are major causes of damage to the periodontal ligament (PDL). Repairing the native conditions of the PDL is essential for the stability of the tissue and its interfaces. Bioprinting periodontal ligament stem cells (PDLSCs) is an interesting approach to guide the regeneration of PDL and interfacial integration. Herein, a collagen-based bioink mimicking the native extracellular matrix conditions and carrying PDLSCs was tested to guide the periodontal ligament organization. The bioink was tested at two different concentrations (10 and 15 mg/mL) and characterized by swelling and degradation, microstructural organization, and rheological properties. The biological properties were assessed after loading PDLSCs into bioinks for bioprinting. The characterization was performed through cell viability, alizarin red assay, and expression for ALP, COL1A1, RUNX2, and OCN. The in vivo biocompatibility of the PDLSC-laden bioinks was verified using subcutaneous implantation in mice. Later, the ability of the bioprinted PDLSC-laden bioinks on dental root fragments to form PDL was also investigated in vivo in mice for 4 and 10 weeks. The bioinks demonstrated typical shear-thinning behavior, a porous microstructure, and stable swelling and degradation characteristics. Both concentrations were printable and provided suitable conditions for a high cell survival, proliferation, and differentiation. PDLSC-laden bioinks demonstrated biocompatibility in vivo, and the bioprinted scaffolds on the root surface evidenced PDLSC alignment, organization, and PDLSC migration to the root surface. The versatility of collagen-based bioinks provides native ECM conditions for PDLSC proliferation, alignment, organization, and differentiation, with translational applications in bioprinting scaffolds for PDL regeneration.
Collapse
Affiliation(s)
- Isaac J de Souza Araújo
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Rachel S Perkins
- Department of Orthopaedic Surgery and Biomedical Engineering, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Mohamed Moustafa Ibrahim
- Department of Ophthalmology, Hamilton Eye Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - George T-J Huang
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Endodontics, The University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Wenjing Zhang
- Department of Genetics, Genomics & Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
4
|
Arslan H, Davuluri A, Nguyen HH, So BR, Lee J, Jeon J, Yum K. 3D Bioprinting Using Universal Fugitive Network Bioinks. ACS APPLIED BIO MATERIALS 2024; 7:7040-7050. [PMID: 39291381 DOI: 10.1021/acsabm.4c01220] [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: 09/19/2024]
Abstract
Three-dimensional (3D) bioprinting has emerged with potential for creating functional 3D tissues with customized geometries. However, the limited availability of bioinks capable of printing 3D structures with both high-shape fidelity and desired biological environments for encapsulated cells remains a key challenge. Here, we present a 3D bioprinting approach that uses universal fugitive network bioinks prepared by loading cells and hydrogel precursors (bioink base materials) into a 3D printable fugitive carrier. This approach constructs 3D structures of cell-encapsulated hydrogels by printing 3D structures using fugitive network bioinks, followed by cross-linking printed structures and removing the carrier from them. The use of the fugitive carrier decouples the 3D printability of bioinks from the material properties of bioink base materials, making this approach readily applicable to a range of hydrogel systems. The decoupling also enables the design of bioinks for the biological functionality of the final printed constructs without compromising the 3D printability. We demonstrate the generalizable 3D printability by printing self-supporting 3D structures of various hydrogels, including conventionally non-3D printable hydrogels and those with a low polymer content. We conduct preprinting screening of bioink base materials through 3D cell culture to select bioinks with high cell compatibility. The selected bioinks produce 3D constructs of cell-encapsulated hydrogels with both high-shape fidelity and high cell viability and proliferation. The universal fugitive network bioink platform could significantly expand 3D printable bioinks with customizable biological functionalities and the adoption of 3D bioprinting in diverse research and applied settings.
Collapse
Affiliation(s)
- Hakan Arslan
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Aneela Davuluri
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Hiep H Nguyen
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Byung Ran So
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Juhyun Lee
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Junha Jeon
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Kyungsuk Yum
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| |
Collapse
|
5
|
Kumi M, Chen T, Zhang Z, Wang A, Li G, Hou Z, Cheng T, Wang J, Wang T, Li P. Integration of Hydrogels and 3D Bioprinting Technologies for Chronic Wound Healing Management. ACS Biomater Sci Eng 2024; 10:5995-6016. [PMID: 39228365 DOI: 10.1021/acsbiomaterials.4c00957] [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: 09/05/2024]
Abstract
The integration of hydrogel-based bioinks with 3D bioprinting technologies presents an innovative approach to chronic wound management, which is particularly challenging to treat because of its multifactorial nature and high risk of complications. Using precise deposition techniques, 3D bioprinting significantly alters traditional wound care paradigms by enabling the fabrication of patient-specific wound dressings that imitate natural tissue properties. Hydrogels are notably beneficial for these applications because of their abundant water content and mechanical properties, which promote cell viability and pathophysiological processes of wound healing, such as re-epithelialization and angiogenesis. This article reviews key 3D printing technologies and their significance in enhancing the structural and functional outcomes of wound-care solutions. Challenges in bioink viscosity, cell viability, and printability are addressed, along with discussions on the cross-linking and mechanical stability of the constructs. The potential of 3D bioprinting to revolutionize chronic wound management rests on its capacity to generate remedies that expedite healing and minimize infection risks. Nevertheless, further studies and clinical trials are necessary to advance these therapies from laboratory to clinical use.
Collapse
Affiliation(s)
- Moses Kumi
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Tianyi Chen
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Zhengheng Zhang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - An Wang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Gangfeng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Zishuo Hou
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Tian Cheng
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Junjie Wang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Tengjiao Wang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
- Chongqing Innovation Center, Northwestern Polytechnical University, Chongqing 401135, P. R. China
- School of Flexible Electronics, Henan Institute of Flexible Electronics (HIFE), Henan University, 379 Mingli Road, Zhengzhou 450046, P. R. China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
- School of Flexible Electronics, Henan Institute of Flexible Electronics (HIFE), Henan University, 379 Mingli Road, Zhengzhou 450046, P. R. China
| |
Collapse
|
6
|
Buchholz MB, Scheerman DI, Levato R, Wehrens EJ, Rios AC. Human breast tissue engineering in health and disease. EMBO Mol Med 2024; 16:2299-2321. [PMID: 39179741 PMCID: PMC11473723 DOI: 10.1038/s44321-024-00112-3] [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: 03/22/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 08/26/2024] Open
Abstract
The human mammary gland represents a highly organized and dynamic tissue, uniquely characterized by postnatal developmental cycles. During pregnancy and lactation, it undergoes extensive hormone-stimulated architectural remodeling, culminating in the formation of specialized structures for milk production to nourish offspring. Moreover, it carries significant health implications, due to the high prevalence of breast cancer. Therefore, gaining insight into the unique biology of the mammary gland can have implications for managing breast cancer and promoting the well-being of both women and infants. Tissue engineering techniques hold promise to narrow the translational gap between existing breast models and clinical outcomes. Here, we provide an overview of the current landscape of breast tissue engineering, outline key requirements, and the challenges to overcome for achieving more predictive human breast models. We propose methods to validate breast function and highlight preclinical applications for improved understanding and targeting of breast cancer. Beyond mammary gland physiology, representative human breast models can offer new insight into stem cell biology and developmental processes that could extend to other organs and clinical contexts.
Collapse
Affiliation(s)
- Maj-Britt Buchholz
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Demi I Scheerman
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Riccardo Levato
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ellen J Wehrens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
7
|
Daly AC. Granular Hydrogels in Biofabrication: Recent Advances and Future Perspectives. Adv Healthc Mater 2024; 13:e2301388. [PMID: 37317658 DOI: 10.1002/adhm.202301388] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/10/2023] [Indexed: 06/16/2023]
Abstract
Granular hydrogels, which are formed by densely packing microgels, are promising materials for bioprinting due to their extrudability, porosity, and modularity. However, the multidimensional parameter space involved in granular hydrogel design makes material optimization challenging. For example, design inputs such as microgel morphology, packing density, or stiffness can influence multiple rheological properties that govern printability and the behavior of encapsulated cells. This review provides an overview of fabrication methods for granular hydrogels, and then examines how important design inputs can influence material properties associated with printability and cellular responses across multiple scales. Recent applications of granular design principles in bioink engineering are described, including the development of granular support hydrogels for embedded printing. Further, the paper provides an overview of how key physical properties of granular hydrogels can influence cellular responses, highlighting the advantages of granular materials for promoting cell and tissue maturation after the printing process. Finally, potential future directions for advancing the design of granular hydrogels for bioprinting are discussed.
Collapse
Affiliation(s)
- Andrew C Daly
- Biomedical Engineering, University of Galway, Galway, H91 TK33, Ireland
- CÚRAM the Science Foundation Ireland Research Centre for Medical Devices, University of Galway, Galway, H91 TK33, Ireland
| |
Collapse
|
8
|
Yao Z, Feng X, Wang Z, Zhan Y, Wu X, Xie W, Wang Z, Zhang G. Techniques and applications in 3D bioprinting with chitosan bio-inks for drug delivery: A review. Int J Biol Macromol 2024; 278:134752. [PMID: 39214837 DOI: 10.1016/j.ijbiomac.2024.134752] [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: 05/01/2024] [Revised: 07/25/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Three-dimensional bioprinting leverages computer-aided design to construct tissues and organs with specialized bioinks. A notable biomaterial for this purpose is chitosan, a natural polysaccharide sourced from crustacean exoskeletons. Chitosan's biocompatibility, biodegradability, non-toxicity, and ability to promote cell adhesion and proliferation make it an excellent component for bioinks. Initially, the rheological properties of chitosan presented challenges for its use in bioprinting. Enhancements in its printability and stability were achieved by integrating it with other natural or synthetic polymers, facilitating its successful application in bioprinting. Chitosan-based bioinks are particularly promising for controlled drug delivery. Incorporating pharmaceuticals directly into the bioink enables the printed structures to serve as localized, sustained-release systems. This approach offers multiple advantages, including precise drug delivery to targeted disease sites, increased therapeutic efficiency, and reduced systemic side effects. Moreover, bioprinting allows for the customization of drug delivery mechanisms to meet individual patient requirements. Although there have been considerable advancements, the use of chitosan-based bioinks in drug delivery is still an emerging field. This review highlights chitosan's essential role in both systemic and localized drug delivery, underscoring its significance and discussing ongoing trends in its application for pharmaceutical purposes.
Collapse
Affiliation(s)
- Zhaomin Yao
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110167, China
| | - Xin Feng
- School of Information and Control Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin, 130011, China
| | - Zheling Wang
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Ying Zhan
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110167, China
| | - Xiaodan Wu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110167, China
| | - Weiming Xie
- School of Information and Control Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin, 130011, China
| | - Zhiguo Wang
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110167, China.
| | - Guoxu Zhang
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110167, China.
| |
Collapse
|
9
|
Wang N, Hu W, Jiang H, Jiang D, Wang L. A portable micro-nanochannel bio-3D printed liver microtissue biosensor for DON detection. Biosens Bioelectron 2024; 267:116810. [PMID: 39357492 DOI: 10.1016/j.bios.2024.116810] [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: 05/29/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024]
Abstract
We investigated a portable micro-nanochannel biosensor 3D-printed liver microtissues for rapid and sensitive deoxynivalenol (DON) detection. The screen-printed carbon electrode (SPCE) was modified with nanoporous anodic aluminum oxide (AAO), gold nanoparticles (AuNPs), and cytochrome C oxidase (COx) to enhance sensor performance. Gelatin methacrylate hydrogel, combined with hepatocellular carcinoma cells, formed the bioink for 3D printing. Liver microtissues were prepared through standardized and high-throughput techniques via bio-3D printing technology. These microtissues were immobilized onto modified electrodes to fabricate liver microtissue sensors. The peak current of this biosensor was positively correlated with DON concentration, as determined by cyclic voltammetry (CV), within a linear detection range of 2∼40 μg/mL. The standard curve equation is denoted by ICV(μA) = = 18.76956 + 0.03107CDON(μg/mL), with a correlation coefficient R2 was 0.99471(n=3). A minimum detection limit of 1.229 μg/mL was calculated from the formula, indicating the successful construction of a portable micro-nanochannel bio-3D printed liver microtissue biosensor. It provides innovative ideas for developing rapid and convenient instrumentation to detect mycotoxin hazards after grain production. It also holds significant potential for application in the prediction and assessment of post-production quality changes in grain.
Collapse
Affiliation(s)
- Nanwei Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China
| | - Wei Hu
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China; Harbin University of Commerce, Harbin, 150028, China
| | - Hui Jiang
- Nanjing Institute for Food and Drug Control, Nanjing, Jiangsu, 211198, China
| | - Donglei Jiang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China.
| | - Lifeng Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
10
|
Li S, Zhang H, Sun L, Zhang X, Guo M, Liu J, Wang W, Zhao N. 4D printing of biological macromolecules employing handheld bioprinters for in situ wound healing applications. Int J Biol Macromol 2024; 280:135999. [PMID: 39326614 DOI: 10.1016/j.ijbiomac.2024.135999] [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: 07/02/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
In situ bioprinting may be preferred over standard in vitro bioprinting in specific cases when de novo tissues are to be created directly on the appropriate anatomical region in the live organism, employing the body as a bioreactor. So far, few efforts have been made to create in situ tissues that can be safely halted and immobilized during printing in preclinical live animals. However, the technique has to be improved significantly in order to manufacture complex tissues in situ, which may be attainable in the future thanks to multidisciplinary advances in tissue engineering. Thanks to the biological macromolecules, natural and synthetic hydrogels and polymers are among the most used biomaterials in in situ bioprinting procedure. Bioprinters, which encounter multiple challenges, including cross-linking the printed structure, adjusting the rheology parameters, and printing various constructs. The introduction of handheld 3D and 4D bioprinters might potentially overcome the difficulties and problems associated with using traditional bioprinters. Studies showed that this technique could be efficient in wound healing and skin tissue regeneration. This study aims to analyze the benefits and difficulties associated with materials in situ 4D printing via handheld bioprinters.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Otolaryngology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China
| | - Hongyang Zhang
- Department of Otolaryngology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China
| | - Lei Sun
- Department of Thoracic surgery, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China
| | - Xinyue Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China
| | - Meiqi Guo
- China Medical University, Shenyang, 110122, Liaoning, China
| | - Jingyang Liu
- China Medical University, Shenyang, 110122, Liaoning, China
| | - Wei Wang
- Department of Otolaryngology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China.
| | - Ning Zhao
- Department of Otolaryngology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China.
| |
Collapse
|
11
|
Kang R, Wu J, Cheng R, Li M, Sang L, Zhang H, Sang S. 3D bioprinting technology and equipment based on microvalve control. Biotechnol Bioeng 2024. [PMID: 39289816 DOI: 10.1002/bit.28850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/26/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
3D bioprinting technology is widely used in biomedical fields such as tissue regeneration and constructing pathological model. The prevailing printing technique is extrusion-based bioprinting. In this printing method, the bioink needs to meet both printability and functionality, which are often conflicting requirements. Therefore, this study has developed an innovative microvalve-based equipment, incorporating components such as pressure control, a three-dimensional motion platform, and microvalve. Here, we present a droplet-based method for constructing complex three-dimensional structures. By leveraging the rapid switching characteristics of the microvalve, this equipment can achieve precise printing of bio-materials with viscosities as low as 10mPa·s, significantly expanding the biofabrication window for bioinks. This technology is of great significance for 3D bioprinting in tissue engineering and lays a solid foundation for the construction of complex artificial organ tissues.
Collapse
Affiliation(s)
- Rihui Kang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan, China
- Shanxi Research Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan, China
| | - Jiaxing Wu
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan, China
- Shanxi Research Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan, China
| | - Rong Cheng
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan, China
- Shanxi Research Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan, China
| | - Meng Li
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan, China
- Shanxi Research Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan, China
| | - Luxiao Sang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Hulin Zhang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Shengbo Sang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan, China
| |
Collapse
|
12
|
Kim DH, Han SG, Lim SJ, Hong SJ, Kwon HC, Jung HS, Han SG. Comparison of Soy and Pea Protein for Cultured Meat Scaffolds: Evaluating Gelation, Physical Properties, and Cell Adhesion. Food Sci Anim Resour 2024; 44:1108-1125. [PMID: 39246534 PMCID: PMC11377198 DOI: 10.5851/kosfa.2024.e46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/08/2024] [Accepted: 06/10/2024] [Indexed: 09/10/2024] Open
Abstract
Cultured meat is under investigation as an environmentally sustainable substitute for conventional animal-derived meat. Employing a scaffolding technique is one approach to developing cultured meat products. The objective of this research was to compare soy and pea protein in the production of hydrogel scaffolds intended for cultured meat. We examined the gelation process, physical characteristics, and the ability of scaffolds to facilitate cell adhesion using mesenchymal stem cells derived from porcine adipose tissue (ADSCs). The combination of soy and pea proteins with agarose and agar powders was found to generate solid hydrogels with a porous structure. Soy protein-based scaffolds exhibited a higher water absorption rate, whereas scaffolds containing agarose had a higher compressive strength. Based on Fourier transform infrared spectroscopy analysis, the number of hydrophobic interactions increased between proteins and polysaccharides in the scaffolds containing pea proteins. All scaffolds were nontoxic toward ADSCs, and soy protein-based scaffolds displayed higher cell adhesion and proliferation properties. Overall, the soy protein-agarose scaffold was found to be optimal for cultured meat production.
Collapse
Affiliation(s)
- Do Hyun Kim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Seo Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Su Jin Lim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Seong Joon Hong
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Hyuk Cheol Kwon
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Hyun Su Jung
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
13
|
Hasan MM, Ahmad A, Akter MZ, Choi YJ, Yi HG. Bioinks for bioprinting using plant-derived biomaterials. Biofabrication 2024; 16:042004. [PMID: 39079554 DOI: 10.1088/1758-5090/ad6932] [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: 09/26/2023] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
Three-dimensional (3D) bioprinting has revolutionized tissue engineering by enabling the fabrication of complex and functional human tissues and organs. An essential component of successful 3D bioprinting is the selection of an appropriate bioink capable of supporting cell proliferation and viability. Plant-derived biomaterials, because of their abundance, biocompatibility, and tunable properties, hold promise as bioink sources, thus offering advantages over animal-derived biomaterials, which carry immunogenic concerns. This comprehensive review explores and analyzes the potential of plant-derived biomaterials as bioinks for 3D bioprinting of human tissues. Modification and optimization of these materials to enhance printability and biological functionality are discussed. Furthermore, cancer research and drug testing applications of the use of plant-based biomaterials in bioprinting various human tissues such as bone, cartilage, skin, and vascular tissues are described. Challenges and limitations, including mechanical integrity, cell viability, resolution, and regulatory concerns, along with potential strategies to overcome them, are discussed. Additionally, this review provides insights into the potential use of plant-based decellularized ECM (dECM) as bioinks, future prospects, and emerging trends in the use of plant-derived biomaterials for 3D bioprinting applications. The potential of plant-derived biomaterials as bioinks for 3D bioprinting of human tissues is highlighted herein. However, further research is necessary to optimize their processing, standardize their properties, and evaluate their long-termin vivoperformance. Continued advancements in plant-derived biomaterials have the potential to revolutionize tissue engineering and facilitate the development of functional and regenerative therapies for diverse clinical applications.
Collapse
Affiliation(s)
- Md Mehedee Hasan
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences (CALS), Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ashfaq Ahmad
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences (CALS), Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, Republic of Korea
| | - Mst Zobaida Akter
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences (CALS), Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, Republic of Korea
| | - Yeong-Jin Choi
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon 51508, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences (CALS), Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
14
|
Soliman BG, Chin IL, Li Y, Ishii M, Ho MH, Doan VK, Cox TR, Wang PY, Lindberg GCJ, Zhang YS, Woodfield TBF, Choi YS, Lim KS. Droplet-based microfluidics for engineering shape-controlled hydrogels with stiffness gradient. Biofabrication 2024; 16:045026. [PMID: 39121873 DOI: 10.1088/1758-5090/ad6d8e] [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: 03/10/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Current biofabrication strategies are limited in their ability to replicate native shape-to-function relationships, that are dependent on adequate biomimicry of macroscale shape as well as size and microscale spatial heterogeneity, within cell-laden hydrogels. In this study, a novel diffusion-based microfluidics platform is presented that meets these needs in a two-step process. In the first step, a hydrogel-precursor solution is dispersed into a continuous oil phase within the microfluidics tubing. By adjusting the dispersed and oil phase flow rates, the physical architecture of hydrogel-precursor phases can be adjusted to generate spherical and plug-like structures, as well as continuous meter-long hydrogel-precursor phases (up to 1.75 m). The second step involves the controlled introduction a small molecule-containing aqueous phase through a T-shaped tube connector to enable controlled small molecule diffusion across the interface of the aqueous phase and hydrogel-precursor. Application of this system is demonstrated by diffusing co-initiator sodium persulfate (SPS) into hydrogel-precursor solutions, where the controlled SPS diffusion into the hydrogel-precursor and subsequent photo-polymerization allows for the formation of unique radial stiffness patterns across the shape- and size-controlled hydrogels, as well as allowing the formation of hollow hydrogels with controllable internal architectures. Mesenchymal stromal cells are successfully encapsulated within hollow hydrogels and hydrogels containing radial stiffness gradient and found to respond to the heterogeneity in stiffness through the yes-associated protein mechano-regulator. Finally, breast cancer cells are found to phenotypically switch in response to stiffness gradients, causing a shift in their ability to aggregate, which may have implications for metastasis. The diffusion-based microfluidics thus finds application mimicking native shape-to-function relationship in the context of tissue engineering and provides a platform to further study the roles of micro- and macroscale architectural features that exist within native tissues.
Collapse
Affiliation(s)
- Bram G Soliman
- Light Activated Biomaterials (LAB) Group, University of Otago, Christchurch 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
- School of Material Science and Engineering, University of New South Wales, Sydney 2052, Australia
| | - Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth 6009, Australia
| | - Yiwei Li
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| | - Melissa Ishii
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
| | - Minh Hieu Ho
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| | - Vinh Khanh Doan
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| | - Thomas R Cox
- The Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Peng Yuan Wang
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 32500, People's Republic of China
| | - Gabriella C J Lindberg
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, United States of America
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth 6009, Australia
| | - Khoon S Lim
- Light Activated Biomaterials (LAB) Group, University of Otago, Christchurch 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
15
|
Ng WL, Goh GL, Goh GD, Ten JSJ, Yeong WY. Progress and Opportunities for Machine Learning in Materials and Processes of Additive Manufacturing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310006. [PMID: 38456831 DOI: 10.1002/adma.202310006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 03/01/2024] [Indexed: 03/09/2024]
Abstract
In recent years, there has been widespread adoption of machine learning (ML) technologies to unravel intricate relationships among diverse parameters in various additive manufacturing (AM) techniques. These ML models excel at recognizing complex patterns from extensive, well-curated datasets, thereby unveiling latent knowledge crucial for informed decision-making during the AM process. The collaborative synergy between ML and AM holds the potential to revolutionize the design and production of AM-printed parts. This review delves into the challenges and opportunities emerging at the intersection of these two dynamic fields. It provides a comprehensive analysis of the publication landscape for ML-related research in the field of AM, explores common ML applications in AM research (such as quality control, process optimization, design optimization, microstructure analysis, and material formulation), and concludes by presenting an outlook that underscores the utilization of advanced ML models, the development of emerging sensors, and ML applications in emerging AM-related fields. Notably, ML has garnered increased attention in AM due to its superior performance across various AM-related applications. It is envisioned that the integration of ML into AM processes will significantly enhance 3D printing capabilities across diverse AM-related research areas.
Collapse
Affiliation(s)
- Wei Long Ng
- Singapore Centre for 3D Printing, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Guo Liang Goh
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Ave, Singapore, 639798, Singapore
| | - Guo Dong Goh
- Singapore Institute of Manufacturing Technology (SIMTech), Agency for Science, Technology and Research (A*STAR), 5 CleanTech Loop #01-01, Singapore, 636732, Singapore
| | - Jyi Sheuan Jason Ten
- Singapore Institute of Manufacturing Technology (SIMTech), Agency for Science, Technology and Research (A*STAR), 5 CleanTech Loop #01-01, Singapore, 636732, Singapore
| | - Wai Yee Yeong
- Singapore Centre for 3D Printing, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Ave, Singapore, 639798, Singapore
| |
Collapse
|
16
|
Grottkau BE, Hui Z, Pang Y. Cellular Patterning Alone Using Bioprinting Regenerates Articular Cartilage Through Native-Like Cartilagenesis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308694. [PMID: 38763898 DOI: 10.1002/smll.202308694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 04/12/2024] [Indexed: 05/21/2024]
Abstract
Few studies have proved that bioprinting itself helps recapitulate native tissue functions mainly because the bioprinted macro shape can rarely, if ever, influence cell function. This can be more problematic in bioprinting cartilage, generally considered more challenging to engineer. Here a new method is shown to micro-pattern chondrocytes within bioprinted sub-millimeter micro tissues, denoted as patterned micro-articular-cartilages tissues (PA-MCTs). Under the sole influence of bioprinted cellular patterns. A pattern scoring system is developed after over 600 bioprinted cellular patterns are analyzed. The top-scored pattern mimics that of the isogenous group in native articular cartilage. Under the sole influence of this pattern during PA-MCTs bio-assembling into macro-cartilage and repairing cartilage defects, chondrogenic cell phenotype is preserved, and cartilagenesis is initiated and maintained. Neocartilage tissues from individual and assembled PA-MCTs are comparable to native articular cartilage and superior to cartilage bioprinted with homogeneously distributed cells in morphology, biochemical components, cartilage-specific protein and gene expression, mechanical properties, integration with host tissues, zonation forming and stem cell chondrogenesis. PA-MCTs can also be used as osteoarthritic and healthy cartilage models for therapeutic drug screening and cartilage development studies. This cellular patterning technique can pave a new way for bioprinting to recapitulate native tissue functions via tissue genesis.
Collapse
Affiliation(s)
- Brian E Grottkau
- The Laboratory for Therapeutic 3D Bioprinting, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhixin Hui
- The Laboratory for Therapeutic 3D Bioprinting, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Yonggang Pang
- The Laboratory for Therapeutic 3D Bioprinting, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
17
|
Wang Y, Zhang C, Cheng J, Yan T, He Q, Huang D, Liu J, Wang Z. Cutting-Edge Biomaterials in Intervertebral Disc Degeneration Tissue Engineering. Pharmaceutics 2024; 16:979. [PMID: 39204324 PMCID: PMC11359550 DOI: 10.3390/pharmaceutics16080979] [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: 05/31/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) stands as the foremost contributor to low back pain (LBP), imposing a substantial weight on the world economy. Traditional treatment modalities encompass both conservative approaches and surgical interventions; however, the former falls short in halting IVDD progression, while the latter carries inherent risks. Hence, the quest for an efficacious method to reverse IVDD onset is paramount. Biomaterial delivery systems, exemplified by hydrogels, microspheres, and microneedles, renowned for their exceptional biocompatibility, biodegradability, biological efficacy, and mechanical attributes, have found widespread application in bone, cartilage, and various tissue engineering endeavors. Consequently, IVD tissue engineering has emerged as a burgeoning field of interest. This paper succinctly introduces the intervertebral disc (IVD) structure and the pathophysiology of IVDD, meticulously classifies biomaterials for IVD repair, and reviews recent advances in the field. Particularly, the strengths and weaknesses of biomaterials in IVD tissue engineering are emphasized, and potential avenues for future research are suggested.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Chuyue Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Junyao Cheng
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Taoxu Yan
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Qing He
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China; (Q.H.); (D.H.)
| | - Da Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China; (Q.H.); (D.H.)
| | - Jianheng Liu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Zheng Wang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| |
Collapse
|
18
|
Terriac L, Helesbeux JJ, Maugars Y, Guicheux J, Tibbitt MW, Delplace V. Boronate Ester Hydrogels for Biomedical Applications: Challenges and Opportunities. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2024; 36:6674-6695. [PMID: 39070669 PMCID: PMC11270748 DOI: 10.1021/acs.chemmater.4c00507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 07/30/2024]
Abstract
Boronate ester (BE) hydrogels are increasingly used for biomedical applications. The dynamic nature of these molecular networks enables bond rearrangement, which is associated with viscoelasticity, injectability, printability, and self-healing, among other properties. BEs are also sensitive to pH, redox reactions, and the presence of sugars, which is useful for the design of stimuli-responsive materials. Together, BE hydrogels are interesting scaffolds for use in drug delivery, 3D cell culture, and biofabrication. However, designing stable BE hydrogels at physiological pH (≈7.4) remains a challenge, which is hindering their development and biomedical application. In this context, advanced chemical insights into BE chemistry are being used to design new molecular solutions for material fabrication. This review article summarizes the state of the art in BE hydrogel design for biomedical applications with a focus on the materials chemistry of this class of materials. First, we discuss updated knowledge in BE chemistry including details on the molecular mechanisms associated with BE formation and breakage. Then, we discuss BE hydrogel formation at physiological pH, with an overview of the main systems reported to date along with new perspectives. A last section covers several prominent biomedical applications of BE hydrogels, including drug delivery, 3D cell culture, and bioprinting, with critical insights on the design relevance, limitations and potential.
Collapse
Affiliation(s)
- Léa Terriac
- Nantes
Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton,
RMeS, UMR 1229, F-44000 Nantes, France
| | | | - Yves Maugars
- Nantes
Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton,
RMeS, UMR 1229, F-44000 Nantes, France
| | - Jérôme Guicheux
- Nantes
Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton,
RMeS, UMR 1229, F-44000 Nantes, France
| | - Mark W. Tibbitt
- Macromolecular
Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Vianney Delplace
- Nantes
Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton,
RMeS, UMR 1229, F-44000 Nantes, France
| |
Collapse
|
19
|
Xiao L, Liu H, Huang H, Wu S, Xue L, Geng Z, Cai L, Yan F. 3D nanofiber scaffolds from 2D electrospun membranes boost cell penetration and positive host response for regenerative medicine. J Nanobiotechnology 2024; 22:322. [PMID: 38849858 PMCID: PMC11162076 DOI: 10.1186/s12951-024-02578-2] [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: 11/10/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
The ideal tissue engineering scaffold should facilitate rapid cell infiltration and provide an optimal immune microenvironment during interactions with the host. Electrospinning can produce two-dimensional (2D) membranes mimicking the extracellular matrix. However, their dense structure hinders cell penetration, and their thin form restricts scaffold utility. In this study, latticed hydrogels were three-dimensional (3D) printed onto electrospun membranes. This technique allowed for layer-by-layer assembly of the membranes into 3D scaffolds, which maintained their resilience impressively under both dry and wet conditions. We assessed the cellular and host responses of these 3D nanofiber scaffolds by comparing random membranes and mesh-like membranes with three different mesh sizes (250, 500, and 750 μm). It was found that scaffolds with a mesh size of 500 μm were superior for M2 macrophage phenotype polarization, vascularization, and matrix deposition. Furthermore, it was confirmed by subsequent experiments such as RNA sequencing that the mesh-like topology may promote polarization to the M2 phenotype by affecting the PI3K/AKT pathway. In conclusion, our work offers a novel method for transforming 2D nanofiber membranes into 3D scaffolds. This method boasts flexibility, allowing for the use of varied electrospun membranes and hydrogels in terms of structure and composition. It has vast potential in tissue repair and regeneration.
Collapse
Affiliation(s)
- Lingfei Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Huifan Liu
- Department of Anesthesiology, Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Huayi Huang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shujuan Wu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Longjian Xue
- The Institute of Technological Science, School of Power and Mechanical Engineering, Wuhan University, Wuhan, 430072, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Feifei Yan
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
20
|
Huang Y, Hu W, Xu K, Dan R, Tan S, Shu Z, Li X, Liu H, Fan C, Xing M, Yang S. Plant mucus-derived microgels: Blood-triggered gelation and strong hemostatic adhesion. Biomaterials 2024; 307:122535. [PMID: 38518590 DOI: 10.1016/j.biomaterials.2024.122535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/24/2024]
Abstract
Arrest of bleeding usually applies clotting agents to trigger coagulation procedures or adhesives to interrupt blood flow through sealing the vessel; however, the efficiency is compromised. Here, we propose a concept of integration of hemostasis and adhesion via yam mucus's microgels. The mucus microgels exhibit attractive attributes of hydrogel with uniform size and shape. Their shear-thinning, self-healing and strong adhesion make them feasible as injectable bioadhesion. Exceptionally, the blood can trigger the microgels' gelation with the outcome of super extensibility, which leads to the microgels a strong hemostatic agent. We also found a tight gel adhesive layer formed upon microgels' contacting the blood on the tissue, where there is the coagulation factor XIII triggered to form a dense three-dimensional fibrin meshwork. The generated structures show that the microgels look like hard balls in the dispersed phase into the blood-produced fibrin mesh of a soft net phase. Both phases work together for a super-extension gel. We demonstrated the microgels' fast adhesion and hemostasis in the livers and hearts of rabbits and mini pigs. The microgels also promoted wound healing with good biocompatibility and biodegradability.
Collapse
Affiliation(s)
- Yu Huang
- Department of Gastroenterology, Xinqiao Hospital, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Weichao Hu
- Department of Gastroenterology, Xinqiao Hospital, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Kaige Xu
- Department of Mechanical Engineering, University of Manitoba, Winnipeg MB, R3T 2N2, Manitoba, Canada
| | - Ruijue Dan
- Department of Gastroenterology, Xinqiao Hospital, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Shali Tan
- Department of Gastroenterology, Xinqiao Hospital, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Zhenzhen Shu
- Department of Gastroenterology, Xinqiao Hospital, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Xin Li
- Department of Gastroenterology, Xinqiao Hospital, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Hangzong Liu
- Department of Gastroenterology, Xinqiao Hospital, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Chaoqiang Fan
- Department of Gastroenterology, Xinqiao Hospital, NO.183, Xinqiao Street, Chongqing, 400037, China; Chongqing Municipality Clinical Research Center for Gastroenterology, Chongqing, 400037, China.
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg MB, R3T 2N2, Manitoba, Canada.
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, NO.183, Xinqiao Street, Chongqing, 400037, China; Chongqing Municipality Clinical Research Center for Gastroenterology, Chongqing, 400037, China.
| |
Collapse
|
21
|
Almalla A, Elomaa L, Fribiczer N, Landes T, Tang P, Mahfouz Z, Koksch B, Hillebrandt KH, Sauer IM, Heinemann D, Seiffert S, Weinhart M. Chemistry matters: A side-by-side comparison of two chemically distinct methacryloylated dECM bioresins for vat photopolymerization. BIOMATERIALS ADVANCES 2024; 160:213850. [PMID: 38626580 DOI: 10.1016/j.bioadv.2024.213850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 04/18/2024]
Abstract
Decellularized extracellular matrix (dECM) is an excellent natural source for 3D bioprinting materials due to its inherent cell compatibility. In vat photopolymerization, the use of dECM-based bioresins is just emerging, and extensive research is needed to fully exploit their potential. In this study, two distinct methacryloyl-functionalized, photocrosslinkable dECM-based bioresins were prepared from digested porcine liver dECM through functionalization with glycidyl methacrylate (GMA) or conventional methacrylic anhydride (MA) under mild conditions for systematic comparison. Although the chemical modifications did not significantly affect the structural integrity of the dECM proteins, mammalian cells encapsulated in the respective hydrogels performed differently in long-term culture. In either case, photocrosslinking during 3D (bio)printing resulted in transparent, highly swollen, and soft hydrogels with good shape fidelity, excellent biomimetic properties and tunable mechanical properties (~ 0.2-2.5 kPa). Interestingly, at a similar degree of functionalization (DOF ~ 81.5-83.5 %), the dECM-GMA resin showed faster photocrosslinking kinetics in photorheology resulting in lower final stiffness and faster enzymatic biodegradation compared to the dECM-MA gels, yet comparable network homogeneity as assessed via Brillouin imaging. While human hepatic HepaRG cells exhibited comparable cell viability directly after 3D bioprinting within both materials, cell proliferation and spreading were clearly enhanced in the softer dECM-GMA hydrogels at a comparable degree of crosslinking. These differences were attributed to the additional hydrophilicity introduced to dECM via methacryloylation through GMA compared to MA. Due to its excellent printability and cytocompatibility, the functional porcine liver dECM-GMA biomaterial enables the advanced biofabrication of soft 3D tissue analogs using vat photopolymerization-based bioprinting.
Collapse
Affiliation(s)
- Ahed Almalla
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Nora Fribiczer
- Department of Chemistry, Johannes Gutenberg Universität Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Timm Landes
- HOT - Hanover Centre for Optical Technologies, Leibniz Universität Hannover, Nienburger Straße 17, 30167 Hannover, Germany; Institute of Horticultural Productions Systems, Leibniz Universität Hannover, Herrenhäuser Straße 2, 30419 Hannover, Germany; Cluster of Excellence PhoenixD, Leibniz University Hannover, Welfengarten 1a, 30167 Hannover, Germany
| | - Peng Tang
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Zeinab Mahfouz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Beate Koksch
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Karl Herbert Hillebrandt
- Experimental Surgery, Department of Surgery, CCM|CVK, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Charitéplatz 1, 10117 Berlin, Germany; Cluster of Excellence Matters of Activity, Image Space Material funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany's Excellence Strategy - EXC 2025, Germany
| | - Igor Maximilian Sauer
- Experimental Surgery, Department of Surgery, CCM|CVK, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Cluster of Excellence Matters of Activity, Image Space Material funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany's Excellence Strategy - EXC 2025, Germany
| | - Dag Heinemann
- HOT - Hanover Centre for Optical Technologies, Leibniz Universität Hannover, Nienburger Straße 17, 30167 Hannover, Germany; Institute of Horticultural Productions Systems, Leibniz Universität Hannover, Herrenhäuser Straße 2, 30419 Hannover, Germany; Cluster of Excellence PhoenixD, Leibniz University Hannover, Welfengarten 1a, 30167 Hannover, Germany
| | - Sebastian Seiffert
- Department of Chemistry, Johannes Gutenberg Universität Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany; Cluster of Excellence Matters of Activity, Image Space Material funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany's Excellence Strategy - EXC 2025, Germany; Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstr. 3A, 30167 Hannover, Germany.
| |
Collapse
|
22
|
Haidar LL, Bilek M, Akhavan B. Surface Bio-engineered Polymeric Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310876. [PMID: 38396265 DOI: 10.1002/smll.202310876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Surface bio-engineering of polymeric nanoparticles (PNPs) has emerged as a cornerstone in contemporary biomedical research, presenting a transformative avenue that can revolutionize diagnostics, therapies, and drug delivery systems. The approach involves integrating bioactive elements on the surfaces of PNPs, aiming to provide them with functionalities to enable precise, targeted, and favorable interactions with biological components within cellular environments. However, the full potential of surface bio-engineered PNPs in biomedicine is hampered by obstacles, including precise control over surface modifications, stability in biological environments, and lasting targeted interactions with cells or tissues. Concerns like scalability, reproducibility, and long-term safety also impede translation to clinical practice. In this review, these challenges in the context of recent breakthroughs in developing surface-biofunctionalized PNPs for various applications, from biosensing and bioimaging to targeted delivery of therapeutics are discussed. Particular attention is given to bonding mechanisms that underlie the attachment of bioactive moieties to PNP surfaces. The stability and efficacy of surface-bioengineered PNPs are critically reviewed in disease detection, diagnostics, and treatment, both in vitro and in vivo settings. Insights into existing challenges and limitations impeding progress are provided, and a forward-looking discussion on the field's future is presented. The paper concludes with recommendations to accelerate the clinical translation of surface bio-engineered PNPs.
Collapse
Affiliation(s)
- Laura Libnan Haidar
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Marcela Bilek
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Behnam Akhavan
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- School of Engineering, University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute (HMRI), Precision Medicine Program, New Lambton Heights, NSW, 2305, Australia
| |
Collapse
|
23
|
Chrungoo S, Bharadwaj T, Verma D. Nanofibrous polyelectrolyte complex incorporated BSA-alginate composite bioink for 3D bioprinting of bone mimicking constructs. Int J Biol Macromol 2024; 266:131123. [PMID: 38537853 DOI: 10.1016/j.ijbiomac.2024.131123] [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: 09/26/2023] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/01/2024]
Abstract
Although several bioinks have been developed for 3D bioprinting applications, the lack of optimal printability, mechanical properties, and adequate cell response has limited their practical applicability. Therefore, this work reports the development of a composite bioink consisting of bovine serum albumin (BSA), alginate, and self-assembled nanofibrous polyelectrolyte complex aggregates of gelatin and chitosan (PEC-GC). The nanofibrous PEC-GC aggregates were prepared and incorporated into the bioink in varying concentrations (0 % to 3 %). The bioink samples were bioprinted and crosslinked post-printing by calcium chloride. The average nanofiber diameter of PEC-GC was 62 ± 15 nm. It was demonstrated that PEC-GC improves the printability and cellular adhesion of the developed bioink and modulates the swelling ratio, degradation rate, and mechanical properties of the fabricated scaffold. The in vitro results revealed that the bioink with 2 % PEC-GC had the best post-printing cell viability of the encapsulated MG63 osteosarcoma cells and well oragnized stress fibers, indicating enhanced cell adhesion. The cell viability was >90 %, as observed from the MTT assay. The composite bioink also showed osteogenic potential, as confirmed by the estimation of alkaline phosphatase activity and collagen synthesis assay. This study successfully fabricated a high-shape fidelity bioink with potential in bone tissue engineering.
Collapse
Affiliation(s)
- Shreya Chrungoo
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Tanmay Bharadwaj
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Devendra Verma
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India.
| |
Collapse
|
24
|
Kim W, Kim G. Engineered 3D liver-tissue model with minispheroids formed by a bioprinting process supported with in situ electrical stimulation. Bioact Mater 2024; 35:382-400. [PMID: 38379698 PMCID: PMC10876469 DOI: 10.1016/j.bioactmat.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/22/2024] Open
Abstract
Three-dimensional (3D) bioprinting, an effective technique for building cell-laden structures providing native extracellular matrix environments, presents challenges, including inadequate cellular interactions. To address these issues, cell spheroids offer a promising solution for improving their biological functions. Particularly, minispheroids with 50-100 μm diameters exhibit enhanced cellular maturation. We propose a one-step minispheroid-forming bioprinting process incorporating electrical stimulation (E-MS-printing). By stimulating the cells, minispheroids with controlled diameters were generated by manipulating the bioink viscosity and stimulation intensity. To validate its feasibility, E-MS-printing process was applied to fabricate an engineered liver model designed to mimic the hepatic lobule unit. E-MS-printing was employed to print the hepatocyte region, followed by bioprinting the central vein using a core-shell nozzle. The resulting constructs displayed native liver-mimetic structures containing minispheroids, which facilitated improved hepatic cell maturation, functional attributes, and vessel formation. Our results demonstrate a new potential 3D liver model that can replicate native liver tissues.
Collapse
Affiliation(s)
- WonJin Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
| | - GeunHyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
25
|
Soliman BG, Longoni A, Major GS, Lindberg GCJ, Choi YS, Zhang YS, Woodfield TBF, Lim KS. Harnessing Macromolecular Chemistry to Design Hydrogel Micro- and Macro-Environments. Macromol Biosci 2024; 24:e2300457. [PMID: 38035637 DOI: 10.1002/mabi.202300457] [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: 10/07/2023] [Revised: 11/16/2023] [Indexed: 12/02/2023]
Abstract
Cell encapsulation within three-dimensional hydrogels is a promising approach to mimic tissues. However, true biomimicry of the intricate microenvironment, biophysical and biochemical gradients, and the macroscale hierarchical spatial organizations of native tissues is an unmet challenge within tissue engineering. This review provides an overview of the macromolecular chemistries that have been applied toward the design of cell-friendly hydrogels, as well as their application toward controlling biophysical and biochemical bulk and gradient properties of the microenvironment. Furthermore, biofabrication technologies provide the opportunity to simultaneously replicate macroscale features of native tissues. Biofabrication strategies are reviewed in detail with a particular focus on the compatibility of these strategies with the current macromolecular toolkit described for hydrogel design and the challenges associated with their clinical translation. This review identifies that the convergence of the ever-expanding macromolecular toolkit and technological advancements within the field of biofabrication, along with an improved biological understanding, represents a promising strategy toward the successful tissue regeneration.
Collapse
Affiliation(s)
- Bram G Soliman
- School of Materials Science and Engineering, University of New South Wales, Sydney, 2052, Australia
| | - Alessia Longoni
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3584CX, The Netherlands
| | - Gretel S Major
- Department of Orthopedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Gabriella C J Lindberg
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, 6009, Australia
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02115, USA
| | - Tim B F Woodfield
- Department of Orthopedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Khoon S Lim
- Department of Orthopedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
- School of Medical Sciences, University of Sydney, Sydney, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, 2006, Australia
| |
Collapse
|
26
|
Puertas-Bartolomé M, Venegas-Bustos D, Acosta S, Rodríguez-Cabello JC. Contribution of the ELRs to the development of advanced in vitro models. Front Bioeng Biotechnol 2024; 12:1363865. [PMID: 38650751 PMCID: PMC11033926 DOI: 10.3389/fbioe.2024.1363865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Developing in vitro models that accurately mimic the microenvironment of biological structures or processes holds substantial promise for gaining insights into specific biological functions. In the field of tissue engineering and regenerative medicine, in vitro models able to capture the precise structural, topographical, and functional complexity of living tissues, prove to be valuable tools for comprehending disease mechanisms, assessing drug responses, and serving as alternatives or complements to animal testing. The choice of the right biomaterial and fabrication technique for the development of these in vitro models plays an important role in their functionality. In this sense, elastin-like recombinamers (ELRs) have emerged as an important tool for the fabrication of in vitro models overcoming the challenges encountered in natural and synthetic materials due to their intrinsic properties, such as phase transition behavior, tunable biological properties, viscoelasticity, and easy processability. In this review article, we will delve into the use of ELRs for molecular models of intrinsically disordered proteins (IDPs), as well as for the development of in vitro 3D models for regenerative medicine. The easy processability of the ELRs and their rational design has allowed their use for the development of spheroids and organoids, or bioinks for 3D bioprinting. Thus, incorporating ELRs into the toolkit of biomaterials used for the fabrication of in vitro models, represents a transformative step forward in improving the accuracy, efficiency, and functionality of these models, and opening up a wide range of possibilities in combination with advanced biofabrication techniques that remains to be explored.
Collapse
Affiliation(s)
- María Puertas-Bartolomé
- Technical Proteins Nanobiotechnology, S.L. (TPNBT), Valladolid, Spain
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Desiré Venegas-Bustos
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Sergio Acosta
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
27
|
Das S, Jegadeesan JT, Basu B. Gelatin Methacryloyl (GelMA)-Based Biomaterial Inks: Process Science for 3D/4D Printing and Current Status. Biomacromolecules 2024; 25:2156-2221. [PMID: 38507816 DOI: 10.1021/acs.biomac.3c01271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Tissue engineering for injured tissue replacement and regeneration has been a subject of investigation over the last 30 years, and there has been considerable interest in using additive manufacturing to achieve these goals. Despite such efforts, many key questions remain unanswered, particularly in the area of biomaterial selection for these applications as well as quantitative understanding of the process science. The strategic utilization of biological macromolecules provides a versatile approach to meet diverse requirements in 3D printing, such as printability, buildability, and biocompatibility. These molecules play a pivotal role in both physical and chemical cross-linking processes throughout the biofabrication, contributing significantly to the overall success of the 3D printing process. Among the several bioprintable materials, gelatin methacryloyl (GelMA) has been widely utilized for diverse tissue engineering applications, with some degree of success. In this context, this review will discuss the key bioengineering approaches to identify the gelation and cross-linking strategies that are appropriate to control the rheology, printability, and buildability of biomaterial inks. This review will focus on the GelMA as the structural (scaffold) biomaterial for different tissues and as a potential carrier vehicle for the transport of living cells as well as their maintenance and viability in the physiological system. Recognizing the importance of printability toward shape fidelity and biophysical properties, a major focus in this review has been to discuss the qualitative and quantitative impact of the key factors, including microrheological, viscoelastic, gelation, shear thinning properties of biomaterial inks, and printing parameters, in particular, reference to 3D extrusion printing of GelMA-based biomaterial inks. Specifically, we emphasize the different possibilities to regulate mechanical, swelling, biodegradation, and cellular functionalities of GelMA-based bio(material) inks, by hybridization techniques, including different synthetic and natural biopolymers, inorganic nanofillers, and microcarriers. At the close, the potential possibility of the integration of experimental data sets and artificial intelligence/machine learning approaches is emphasized to predict the printability, shape fidelity, or biophysical properties of GelMA bio(material) inks for clinically relevant tissues.
Collapse
Affiliation(s)
- Soumitra Das
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| | | | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| |
Collapse
|
28
|
Moon SH, Park TY, Cha HJ, Yang YJ. Photo-/thermo-responsive bioink for improved printability in extrusion-based bioprinting. Mater Today Bio 2024; 25:100973. [PMID: 38322663 PMCID: PMC10844750 DOI: 10.1016/j.mtbio.2024.100973] [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: 07/12/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Extrusion-based bioprinting has demonstrated significant potential for manufacturing constructs, particularly for 3D cell culture. However, there is a greatly limited number of bioink candidates exploited with extrusion-based bioprinting, as they meet the opposing requirements for printability with indispensable rheological features and for biochemical functionality with desirable microenvironment. In this study, a blend of silk fibroin (SF) and iota-carrageenan (CG) was chosen as a cell-friendly printable material. The SF/CG ink exhibited suitable viscosity and shear-thinning properties, coupled with the rapid sol-gel transition of CG. By employing photo-crosslinking of SF, the printability with Pr value close to 1 and structural integrity of the 3D constructs were significantly improved within a matter of seconds. The printed constructs demonstrated a Young's modulus of approximately 250 kPa, making them suitable for keratinocyte and myoblast cell culture. Furthermore, the high cell adhesiveness and viability (maximum >98%) of the loaded cells underscored the considerable potential of this 3D culture scaffold applied for skin and muscle tissues, which can be easily manipulated using an extrusion-based bioprinter.
Collapse
Affiliation(s)
- Seo Hyung Moon
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, 22212, Republic of Korea
| | - Tae Yoon Park
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Hyung Joon Cha
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
- Medical Science and Engineering, School of Convergence Science and Technology, Pohang University of Science, Pohang, 37673, Republic of Korea
| | - Yun Jung Yang
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, 22212, Republic of Korea
- Inha University Hospital, Incheon, 22332, Republic of Korea
| |
Collapse
|
29
|
Makode S, Maurya S, Niknam SA, Mollocana-Lara E, Jaberi K, Faramarzi N, Tamayol A, Mortazavi M. Three dimensional (bio)printing of blood vessels: from vascularized tissues to functional arteries. Biofabrication 2024; 16:022005. [PMID: 38277671 DOI: 10.1088/1758-5090/ad22ed] [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: 06/28/2023] [Accepted: 01/26/2024] [Indexed: 01/28/2024]
Abstract
Tissue engineering has emerged as a strategy for producing functional tissues and organs to treat diseases and injuries. Many chronic conditions directly or indirectly affect normal blood vessel functioning, necessary for material exchange and transport through the body and within tissue-engineered constructs. The interest in vascular tissue engineering is due to two reasons: (1) functional grafts can be used to replace diseased blood vessels, and (2) engineering effective vasculature within other engineered tissues enables connection with the host's circulatory system, supporting their survival. Among various practices, (bio)printing has emerged as a powerful tool to engineer biomimetic constructs. This has been made possible with precise control of cell deposition and matrix environment along with the advancements in biomaterials. (Bio)printing has been used for both engineering stand-alone vascular grafts as well as vasculature within engineered tissues for regenerative applications. In this review article, we discuss various conditions associated with blood vessels, the need for artificial blood vessels, the anatomy and physiology of different blood vessels, available 3D (bio)printing techniques to fabricate tissue-engineered vascular grafts and vasculature in scaffolds, and the comparison among the different techniques. We conclude our review with a brief discussion about future opportunities in the area of blood vessel tissue engineering.
Collapse
Affiliation(s)
- Shubham Makode
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Satyajit Maurya
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Seyed A Niknam
- Department of Industrial Engineering, Western New England University, Springfield, MA, United States of America
| | - Evelyn Mollocana-Lara
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Kiana Jaberi
- Department of Nutritional Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Faramarzi
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Mehdi Mortazavi
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, United States of America
| |
Collapse
|
30
|
Roppolo I, Caprioli M, Pirri CF, Magdassi S. 3D Printing of Self-Healing Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305537. [PMID: 37877817 DOI: 10.1002/adma.202305537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/11/2023] [Indexed: 10/26/2023]
Abstract
This review article presents a comprehensive overview of the latest advances in the field of 3D printable structures with self-healing properties. Three-dimensional printing (3DP) is a versatile technology that enables the rapid manufacturing of complex geometric structures with precision and functionality not previously attainable. However, the application of 3DP technology is still limited by the availability of materials with customizable properties specifically designed for additive manufacturing. The addition of self-healing properties within 3D printed objects is of high interest as it can improve the performance and lifespan of structural components, and even enable the mimicking of living tissues for biomedical applications, such as organs printing. The review will discuss and analyze the most relevant results reported in recent years in the development of self-healing polymeric materials that can be processed via 3D printing. After introducing the chemical and physical self-healing mechanism that can be exploited, the literature review here reported will focus in particular on printability and repairing performances. At last, actual perspective and possible development field will be critically discussed.
Collapse
Affiliation(s)
- Ignazio Roppolo
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10129, Italy
- Istituto Italiano di Tecnologia, Center for Sustainable Futures @Polito, Via Livorno 60, Turin, 10144, Italy
| | - Matteo Caprioli
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10129, Italy
- Casali Center for Applied Chemistry, Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, 9090145, Israel
| | - Candido F Pirri
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10129, Italy
- Istituto Italiano di Tecnologia, Center for Sustainable Futures @Polito, Via Livorno 60, Turin, 10144, Italy
| | - Shlomo Magdassi
- Casali Center for Applied Chemistry, Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, 9090145, Israel
| |
Collapse
|
31
|
Tuftee C, Alsberg E, Ozbolat IT, Rizwan M. Emerging granular hydrogel bioinks to improve biological function in bioprinted constructs. Trends Biotechnol 2024; 42:339-352. [PMID: 37852853 PMCID: PMC10939978 DOI: 10.1016/j.tibtech.2023.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
Advancements in 3D bioprinting have been hindered by the trade-off between printability and biological functionality. Existing bioinks struggle to meet both requirements simultaneously. However, new types of bioinks composed of densely packed microgels promise to address this challenge. These bioinks possess intrinsic porosity, allowing for cell growth, oxygen and nutrient transport, and better immunomodulatory properties, leading to superior biological functions. In this review, we highlight key trends in the development of these granular bioinks. Using examples, we demonstrate how granular bioinks overcome the trade-off between printability and cell function. Granular bioinks show promise in 3D bioprinting, yet understanding their unique structure-property-function relationships is crucial to fully leverage the transformative capabilities of these new types of bioinks in bioprinting.
Collapse
Affiliation(s)
- Cody Tuftee
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
| | - Eben Alsberg
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL 60612, USA; Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Orthopedic Surgery, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Mechanical & Industrial Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown Veterans Affairs Medical Center (JBVAMC) at Chicago, Chicago, IL 60612, USA
| | - Ibrahim Tarik Ozbolat
- Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA; Engineering Science and Mechanics, Penn State University, University Park, PA 16802, USA; Neurosurgery Department, Penn State University; Hershey, PA 17033, USA; Medical Oncology Department, Cukurova University, Adana 01330, Turkey
| | - Muhammad Rizwan
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA.
| |
Collapse
|
32
|
Comperat L, Chagot L, Massot S, Stachowicz M, Dusserre N, Médina C, Desigaux T, Dupuy J, Fricain J, Oliveira H. Harnessing Human Placental Membrane-Derived Bioinks: Characterization and Applications in Bioprinting and Vasculogenesis. Adv Healthc Mater 2024; 13:e2303370. [PMID: 37942849 PMCID: PMC11469061 DOI: 10.1002/adhm.202303370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Bioprinting applications in the clinical field generate great interest, but developing suitable biomaterial inks for medical settings is a challenge. Placental tissues offer a promising solution due to their abundance, stability, and status as medical waste. They contain basement membrane components, have a clinical history, and support angiogenesis. This study formulates bioinks from two placental tissues, amnion (AM) and chorion (CHO), and compares their unique extracellular matrix (ECM) and growth factor compositions. Rheological properties of the bioinks are evaluated for bioprinting and maturation of human endothelial cells. Both AM and Cho-derived bioinks sustained human endothelial cell viability, proliferation, and maturation, promoting optimal vasculogenesis. These bioinks derived from human sources have significant potential for tissue engineering applications, particularly in supporting vasculogenesis. This research contributes to the advancement of tissue engineering and regenerative medicine, bringing everyone closer to clinically viable bioprinting solutions using placental tissues as valuable biomaterials.
Collapse
Affiliation(s)
- Léo Comperat
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- Inserm U1026Tissue BioengineeringART BioPrintBordeauxF‐33076France
- CHU BordeauxServices d'Odontologie et de Santé BuccaleBordeauxF‐33076France
| | - Lise Chagot
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- Inserm U1026Tissue BioengineeringART BioPrintBordeauxF‐33076France
- CHU BordeauxServices d'Odontologie et de Santé BuccaleBordeauxF‐33076France
| | - Sarah Massot
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- Inserm U1026Tissue BioengineeringART BioPrintBordeauxF‐33076France
- CHU BordeauxServices d'Odontologie et de Santé BuccaleBordeauxF‐33076France
| | - Marie‐Laure Stachowicz
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- Inserm U1026Tissue BioengineeringART BioPrintBordeauxF‐33076France
- CHU BordeauxServices d'Odontologie et de Santé BuccaleBordeauxF‐33076France
| | - Nathalie Dusserre
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- Inserm U1026Tissue BioengineeringART BioPrintBordeauxF‐33076France
- CHU BordeauxServices d'Odontologie et de Santé BuccaleBordeauxF‐33076France
| | - Chantal Médina
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- Inserm U1026Tissue BioengineeringART BioPrintBordeauxF‐33076France
- CHU BordeauxServices d'Odontologie et de Santé BuccaleBordeauxF‐33076France
| | - Théo Desigaux
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- Inserm U1026Tissue BioengineeringART BioPrintBordeauxF‐33076France
- CHU BordeauxServices d'Odontologie et de Santé BuccaleBordeauxF‐33076France
| | - Jean‐William Dupuy
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- University of BordeauxPlateforme ProtéomeBordeaux33000France
| | - Jean‐Christophe Fricain
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- Inserm U1026Tissue BioengineeringART BioPrintBordeauxF‐33076France
- University of BordeauxPlateforme ProtéomeBordeaux33000France
| | - Hugo Oliveira
- University of BordeauxTissue BioengineeringU1026BordeauxF‐33076France
- Inserm U1026Tissue BioengineeringART BioPrintBordeauxF‐33076France
- CHU BordeauxServices d'Odontologie et de Santé BuccaleBordeauxF‐33076France
| |
Collapse
|
33
|
Zhang P, Liu C, Modavi C, Abate A, Chen H. Printhead on a chip: empowering droplet-based bioprinting with microfluidics. Trends Biotechnol 2024; 42:353-368. [PMID: 37777352 DOI: 10.1016/j.tibtech.2023.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/02/2023] [Accepted: 09/11/2023] [Indexed: 10/02/2023]
Abstract
Droplet-based bioprinting has long struggled with the manipulation and dispensation of individual cells from a printhead, hindering the fabrication of artificial cellular structures with high precision. The integration of modern microfluidic modules into the printhead of a bioprinter is emerging as one approach to overcome this bottleneck. This convergence allows for high-accuracy manipulation and spatial control over placement of cells during printing, and enables the fabrication of cell arrays and hierarchical heterogenous microtissues, opening new applications in bioanalysis and high-throughput screening. In this review, we summarize recent developments in the use of microfluidics in droplet printing systems, with consideration of the working principles; present applications extended through microfluidic features; and discuss the future of this technology.
Collapse
Affiliation(s)
- Pengfei Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing, China.
| | - Congying Liu
- School of Mechanical Engineering and Automation, Beihang University, Beijing, China
| | - Cyrus Modavi
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Adam Abate
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Huawei Chen
- School of Mechanical Engineering and Automation, Beihang University, Beijing, China
| |
Collapse
|
34
|
Davern JW, Hipwood L, Bray LJ, Meinert C, Klein TJ. Addition of Laponite to gelatin methacryloyl bioinks improves the rheological properties and printability to create mechanically tailorable cell culture matrices. APL Bioeng 2024; 8:016101. [PMID: 38204454 PMCID: PMC10776181 DOI: 10.1063/5.0166206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Extrusion-based bioprinting has gained widespread popularity in biofabrication due to its ability to assemble cells and biomaterials in precise patterns and form tissue-like constructs. To achieve this, bioinks must have rheological properties suitable for printing while maintaining cytocompatibility. However, many commonly used biomaterials do not meet the rheological requirements and therefore require modification for bioprinting applications. This study demonstrates the incorporation of Laponite-RD (LPN) into gelatin methacryloyl (GelMA) to produce highly customizable bioinks with desired rheological and mechanical properties for extrusion-based bioprinting. Bioink formulations were based on GelMA (5%-15% w/v) and LPN (0%-4% w/v), and a comprehensive rheological design was applied to evaluate key rheological properties necessary for extrusion-based bioprinting. The results showed that GelMA bioinks with LPN (1%-4% w/v) exhibited pronounced shear thinning and viscoelastic behavior, as well as improved thermal stability. Furthermore, a concentration window of 1%-2% (w/v) LPN to 5%-15% GelMA demonstrated enhanced rheological properties and printability required for extrusion-based bioprinting. Construct mechanical properties were highly tunable by varying polymer concentration and photocrosslinking parameters, with Young's moduli ranging from ∼0.2 to 75 kPa. Interestingly, at higher Laponite concentrations, GelMA cross-linking was inhibited, resulting in softer hydrogels. High viability of MCF-7 breast cancer cells was maintained in both free-swelling droplets and printed hydrogels, and metabolically active spheroids formed over 7 days of culture in all conditions. In summary, the addition of 1%-2% (w/v) LPN to gelatin-based bioinks significantly enhanced rheological properties and retained cell viability and proliferation, suggesting its suitability for extrusion-based bioprinting.
Collapse
|
35
|
An C, Zhang S, Xu J, Zhang Y, Dou Z, Shao F, Long C, yang J, Wang H, Liu J. The microparticulate inks for bioprinting applications. Mater Today Bio 2024; 24:100930. [PMID: 38293631 PMCID: PMC10825055 DOI: 10.1016/j.mtbio.2023.100930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/05/2023] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
Three-dimensional (3D) bioprinting has emerged as a groundbreaking technology for fabricating intricate and functional tissue constructs. Central to this technology are the bioinks, which provide structural support and mimic the extracellular environment, which is crucial for cellular executive function. This review summarizes the latest developments in microparticulate inks for 3D bioprinting and presents their inherent challenges. We categorize micro-particulate materials, including polymeric microparticles, tissue-derived microparticles, and bioactive inorganic microparticles, and introduce the microparticle ink formulations, including granular microparticles inks consisting of densely packed microparticles and composite microparticle inks comprising microparticles and interstitial matrix. The formulations of these microparticle inks are also delved into highlighting their capabilities as modular entities in 3D bioprinting. Finally, existing challenges and prospective research trajectories for advancing the design of microparticle inks for bioprinting are discussed.
Collapse
Affiliation(s)
- Chuanfeng An
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Shiying Zhang
- School of Dentistry, Shenzhen University, Shenzhen, 518060, China
| | - Jiqing Xu
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| | - Yujie Zhang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Zhenzhen Dou
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Fei Shao
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Canling Long
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| | - Jianhua yang
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| | - Huanan Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Jia Liu
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| |
Collapse
|
36
|
Guo A, Zhang S, Yang R, Sui C. Enhancing the mechanical strength of 3D printed GelMA for soft tissue engineering applications. Mater Today Bio 2024; 24:100939. [PMID: 38249436 PMCID: PMC10797197 DOI: 10.1016/j.mtbio.2023.100939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Gelatin methacrylate (GelMA) hydrogels have gained significant traction in diverse tissue engineering applications through the utilization of 3D printing technology. As an artificial hydrogel possessing remarkable processability, GelMA has emerged as a pioneering material in the advancement of tissue engineering due to its exceptional biocompatibility and degradability. The integration of 3D printing technology facilitates the precise arrangement of cells and hydrogel materials, thereby enabling the creation of in vitro models that simulate artificial tissues suitable for transplantation. Consequently, the potential applications of GelMA in tissue engineering are further expanded. In tissue engineering applications, the mechanical properties of GelMA are often modified to overcome the hydrogel material's inherent mechanical strength limitations. This review provides a comprehensive overview of recent advancements in enhancing the mechanical properties of GelMA at the monomer, micron, and nano scales. Additionally, the diverse applications of GelMA in soft tissue engineering via 3D printing are emphasized. Furthermore, the potential opportunities and obstacles that GelMA may encounter in the field of tissue engineering are discussed. It is our contention that through ongoing technological progress, GelMA hydrogels with enhanced mechanical strength can be successfully fabricated, leading to the production of superior biological scaffolds with increased efficacy for tissue engineering purposes.
Collapse
Affiliation(s)
- Ao Guo
- Department of Trauma and Pediatric Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 231200, China
| | - Shengting Zhang
- Department of Trauma and Pediatric Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 231200, China
| | - Runhuai Yang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Cong Sui
- Department of Trauma and Pediatric Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 231200, China
| |
Collapse
|
37
|
Salimi S, Graham AM, Wu Y, Song P, Hart LR, Irvine DJ, Wildman RD, Siviour CR, Hayes W. An effective route to the additive manufacturing of a mechanically gradient supramolecular polymer nanocomposite structure. J Mech Behav Biomed Mater 2024; 150:106358. [PMID: 38169206 DOI: 10.1016/j.jmbbm.2023.106358] [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: 06/08/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024]
Abstract
3D Printing techniques are additive methods of fabricating parts directly from computer-aided designs. Whilst the clearest benefit is the realisation of geometrical freedom, multi-material printing allows the introduction of compositional variation and highly tailored product functionality. The paper reports a proof-of-concept additive manufacturing study to deposit a supramolecular polymer and a complementary organic filler to form composites with gradient composition to enable spatial distribution of mechanical properties and functionality by tuning the number of supramolecular interactions. We use a dual-feed extrusion 3D printing process, with feed stocks based on the supramolecular polymer and its organic composite, delivered at ratios predetermined. This allows for production of a graded specimen with varying filler concentration that dictates the mechanical properties. The printed specimen was inspected under dynamic load in a tensile test using digital image correlation to produce full-field deformation maps, which showed clear differences in deformation in regions with varying compositions, corresponding to the designed-in variations. This approach affords a novel method for printing material with graded mechanical properties which are not currently commercially available or easily accessible, however, the method can potentially be directly translated to the generation of biomaterial-based composites featuring gradients of mechanical properties.
Collapse
Affiliation(s)
- Sara Salimi
- Department of Chemistry, University of Reading, Whiteknights, Reading, RG6 6AD, UK; Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W., Hamilton, Ontario, L8S 4M1, Canada
| | - Aaron M Graham
- Department of Engineering Science, University of Oxford, Oxford, OX1 3PJ, UK
| | - Yuyang Wu
- Faculty of Engineering, The University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Peihao Song
- Department of Engineering Science, University of Oxford, Oxford, OX1 3PJ, UK
| | - Lewis R Hart
- Department of Chemistry, University of Reading, Whiteknights, Reading, RG6 6AD, UK
| | - Derek J Irvine
- Faculty of Engineering, The University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Ricky D Wildman
- Faculty of Engineering, The University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Clive R Siviour
- Department of Engineering Science, University of Oxford, Oxford, OX1 3PJ, UK
| | - Wayne Hayes
- Department of Chemistry, University of Reading, Whiteknights, Reading, RG6 6AD, UK.
| |
Collapse
|
38
|
Wang X, Jiang J, Yuan C, Gu L, Zhang X, Yao Y, Shao L. 3D bioprinting of GelMA with enhanced extrusion printability through coupling sacrificial carrageenan. Biomater Sci 2024; 12:738-747. [PMID: 38105707 DOI: 10.1039/d3bm01489d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The potential of 3D bioprinting in tissue engineering and regenerative medicine is enormous, but its implementation is hindered by the reliance on high-strength materials, which restricts the use of low-viscosity, biocompatible materials. Therefore, a major challenge for incorporating 3D bioprinting into tissue engineering is to develop a novel bioprinting platform that can reversibly provide high biological activity materials with a structural support. This study presents a room temperature printing system based on GelMA combined with carrageenan to address this challenge. By leveraging the wide temperature stability range and lubricating properties of carrageenan the room temperature stability of GelMA could be enhanced, as well as creating a solid ink to improve the performance of solid GelMA. Additionally, by utilizing the solubility of carrageenan at 37 °C, it becomes possible to prepare a porous GelMA structure while mimicking the unique extracellular matrix properties of osteocytes through residual carrageenan content and amplifying BMSCs' osteogenesis potential to some extent. Overall, this study provides an innovative technical platform for incorporating a low-viscosity ink into 3D bioprinting and resolves the long-standing contradiction between material printing performance and biocompatibility in bioprinting technology.
Collapse
Affiliation(s)
- Xueping Wang
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, Zhejiang, China.
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Jinhong Jiang
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, Zhejiang, China.
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Chenhui Yuan
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, Zhejiang, China.
- Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Lin Gu
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, Zhejiang, China.
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang, China
| | - XinYu Zhang
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, Zhejiang, China.
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Yudong Yao
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, Zhejiang, China.
| | - Lei Shao
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, Zhejiang, China.
- State Key Laboratory of Fluid Power and Mechatronic Systems, College of Mechanical Engineering, Zhejiang University, Hangzhou 310027, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang, China
| |
Collapse
|
39
|
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
|
40
|
dos Santos AEA, Guadalupe JL, Albergaria JDS, Almeida IA, Moreira AMS, Copola AGL, de Araújo IP, de Paula AM, Neves BRA, Santos JPF, da Silva AB, Jorge EC, Andrade LDO. Random cellulose acetate nanofibers: a breakthrough for cultivated meat production. Front Nutr 2024; 10:1297926. [PMID: 38249608 PMCID: PMC10796801 DOI: 10.3389/fnut.2023.1297926] [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: 09/20/2023] [Accepted: 11/27/2023] [Indexed: 01/23/2024] Open
Abstract
Overcoming the challenge of creating thick, tissue-resembling muscle constructs is paramount in the field of cultivated meat production. This study investigates the remarkable potential of random cellulose acetate nanofibers (CAN) as a transformative scaffold for muscle tissue engineering (MTE), specifically in the context of cultivated meat applications. Through a comparative analysis between random and aligned CAN, utilizing C2C12 and H9c2 myoblasts, we unveil the unparalleled capabilities of random CAN in facilitating muscle differentiation, independent of differentiation media, by exploiting the YAP/TAZ-related mechanotransduction pathway. In addition, we have successfully developed a novel process for stacking cell-loaded CAN sheets, enabling the production of a three-dimensional meat product. C2C12 and H9c2 loaded CAN sheets were stacked (up to four layers) to form a ~300-400 μm thick tissue 2 cm in length, organized in a mesh of uniaxial aligned cells. To further demonstrate the effectiveness of this methodology for cultivated meat purposes, we have generated thick and viable constructs using chicken muscle satellite cells (cSCs) and random CAN. This groundbreaking discovery offers a cost-effective and biomimetic solution for cultivating and differentiating muscle cells, forging a crucial link between tissue engineering and the pursuit of sustainable and affordable cultivated meat production.
Collapse
Affiliation(s)
- Ana Elisa Antunes dos Santos
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jorge Luís Guadalupe
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliano Douglas Silva Albergaria
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Itallo Augusto Almeida
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Amanda Maria Siqueira Moreira
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Aline Gonçalves Lio Copola
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Isabella Paula de Araújo
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Ana Maria de Paula
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bernardo Ruegger Almeida Neves
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - João Paulo Ferreira Santos
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Aline Bruna da Silva
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Erika Cristina Jorge
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luciana de Oliveira Andrade
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
41
|
Monteiro MV, Rocha M, Gaspar VM, Mano JF. Embedded 3D Bioprinting for Engineering Miniaturized In Vitro Tumor Models. Methods Mol Biol 2024; 2764:279-288. [PMID: 38393601 DOI: 10.1007/978-1-0716-3674-9_18] [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: 02/25/2024]
Abstract
Embedded extrusion 3D bioprinting is a rapidly emerging additive manufacturing methodology that provides a precise spatial deposition of synthetic or natural-origin low-viscosity bioinks during the extrusion printing process. Such a strategy has to date unlocked the freeform extrusion biofabrication of complex micro-to-macro-scale living architectures for numerous applications, including tissue engineering and in vitro disease modeling. In this chapter, we describe a suspension bioprinting methodology leveraging a continuous viscoelastic biopolymer supporting bath functionalized with divalent calcium cations to enable a rapid processing of user-defined bioinks toward architecturally complex 3D in vitro tumor models. This highly simple and cost-effective viscoelastic supporting bath enables a full freeform biofabrication of cell-laden 3D tumor-mimetic architectures that exhibit structural stability in culture post-printing. The cytocompatibility of the supporting bath, its ease of removal from biofabricated living constructs, and its adaptability for processing different ECM-mimetic bioinks open avenues for multi-scale fabrication of numerous types of physiomimetic 3D tumor models for preclinical screening of candidate therapeutics.
Collapse
Affiliation(s)
- Maria V Monteiro
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Marta Rocha
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
42
|
Wu Z, Huang D, Wang J, Zhao Y, Sun W, Shen X. Engineering Heterogeneous Tumor Models for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304160. [PMID: 37946674 PMCID: PMC10767453 DOI: 10.1002/advs.202304160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Tumor tissue engineering holds great promise for replicating the physiological and behavioral characteristics of tumors in vitro. Advances in this field have led to new opportunities for studying the tumor microenvironment and exploring potential anti-cancer therapeutics. However, the main obstacle to the widespread adoption of tumor models is the poor understanding and insufficient reconstruction of tumor heterogeneity. In this review, the current progress of engineering heterogeneous tumor models is discussed. First, the major components of tumor heterogeneity are summarized, which encompasses various signaling pathways, cell proliferations, and spatial configurations. Then, contemporary approaches are elucidated in tumor engineering that are guided by fundamental principles of tumor biology, and the potential of a bottom-up approach in tumor engineering is highlighted. Additionally, the characterization approaches and biomedical applications of tumor models are discussed, emphasizing the significant role of engineered tumor models in scientific research and clinical trials. Lastly, the challenges of heterogeneous tumor models in promoting oncology research and tumor therapy are described and key directions for future research are provided.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian Shen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
43
|
Jahani A, Nourbakhsh MS, Ebrahimzadeh MH, Mohammadi M, Yari D, Moradi A. Biomolecules-Loading of 3D-Printed Alginate-Based Scaffolds for Cartilage Tissue Engineering Applications: A Review on Current Status and Future Prospective. THE ARCHIVES OF BONE AND JOINT SURGERY 2024; 12:92-101. [PMID: 38420521 PMCID: PMC10898798 DOI: 10.22038/abjs.2023.73275.3396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 11/11/2023] [Indexed: 03/02/2024]
Abstract
Osteoarthritis (OA) can arise from various factor including trauma, overuse, as well as degeneration resulting from age or disease. The specific treatment options will vary based on the severity of the condition, and the affected joints. Some common treatments for OA include lifestyle modifications, medications, physical therapy, surgery and tissue engineering (TE). For cartilage tissue engineering (CTE), three-dimension (3D) scaffolds are made of biocompatible natural polymers, which allow for the regeneration of new cartilage tissue. An ideal scaffold should possess biological and mechanical properties that closely resemble those of the cartilage tissue, and lead to improved functional of knee. These scaffolds are specifically engineered to serve as replacements for damaged and provide support to the knee joint. 3D-bioprinted scaffolds are made of biocompatible materials natural polymers, which allow for the regeneration of new cartilage. The utilization of 3D bioprinting method has emerged as a novel approach for fabricating scaffolds with optimal properties for CTE applications. This method enables the creation of scaffolds that closely mimic the native cartilage in terms of mechanical characteristics and biological functionality. Alginate, that has the capability to fabricate a cartilage replacement customized for each individual patient. This polymer exhibits hydrophilicity, biocompatibility, and biodegradability, along with shear-thinning properties. These unique properties enable Alginate to be utilized as a bio-ink for 3D bioprinting method. Furthermore, chondrogenesis is the complex process through which cartilage is formed via a series of cellular and molecular signaling. Signaling pathway is as a fundamental mechanism in cartilage formation, enhanced by the incorporation of biomolecules and growth factors that induce the differentiation of stem cells. Accordingly, ongoing review is focusing to promote of 3D bioprinting scaffolds through the utilization of advanced biomolecules-loading of Alginate-based that has the capability to fabricate a cartilage replacement tailored specifically to each patient's unique needs and anatomical requirements.
Collapse
Affiliation(s)
- Afsaneh Jahani
- Faculty of New Sciences and Technologies, Department of Biotechnology , Semnan University, Semnan, Iran
| | - Mohammad Sadegh Nourbakhsh
- These authors have contributed equally as the corresponding author
- Faculty of Materials and Metallurgical Engineering, Semnan University, Semnan, Iran
| | - Mohammad H Ebrahimzadeh
- Bone and Joint Research laboratory, Ghaem Hospital, Mashhad University of Medical Science, Mashhad, Iran
- Orthopedic Research Center, Department of Orthopedic Surgery, Mashhad University of Medical Science, Mashhad, Iran
| | - Marzieh Mohammadi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| | - Davood Yari
- Department of Clinical Biochemistry, Babol University of Medical Science, Babol, Iran
| | - Ali Moradi
- These authors have contributed equally as the corresponding author
- Orthopedic Research Center, Department of Orthopedic Surgery, Mashhad University of Medical Science, Mashhad, Iran
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran
| |
Collapse
|
44
|
Putra NE, Zhou J, Zadpoor AA. Sustainable Sources of Raw Materials for Additive Manufacturing of Bone-Substituting Biomaterials. Adv Healthc Mater 2024; 13:e2301837. [PMID: 37535435 PMCID: PMC11468967 DOI: 10.1002/adhm.202301837] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/12/2023] [Indexed: 08/05/2023]
Abstract
The need for sustainable development has never been more urgent, as the world continues to struggle with environmental challenges, such as climate change, pollution, and dwindling natural resources. The use of renewable and recycled waste materials as a source of raw materials for biomaterials and tissue engineering is a promising avenue for sustainable development. Although tissue engineering has rapidly developed, the challenges associated with fulfilling the increasing demand for bone substitutes and implants remain unresolved, particularly as the global population ages. This review provides an overview of waste materials, such as eggshells, seashells, fish residues, and agricultural biomass, that can be transformed into biomaterials for bone tissue engineering. While the development of recycled metals is in its early stages, the use of probiotics and renewable polymers to improve the biofunctionalities of bone implants is highlighted. Despite the advances of additive manufacturing (AM), studies on AM waste-derived bone-substitutes are limited. It is foreseeable that AM technologies can provide a more sustainable alternative to manufacturing biomaterials and implants. The preliminary results of eggshell and seashell-derived calcium phosphate and rice husk ash-derived silica can likely pave the way for more advanced applications of AM waste-derived biomaterials for sustainably addressing several unmet clinical applications.
Collapse
Affiliation(s)
- Niko E. Putra
- Department of Biomechanical EngineeringFaculty of MechanicalMaritimeand Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Jie Zhou
- Department of Biomechanical EngineeringFaculty of MechanicalMaritimeand Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Amir A. Zadpoor
- Department of Biomechanical EngineeringFaculty of MechanicalMaritimeand Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| |
Collapse
|
45
|
de Ruijter M, Diloksumpan P, Dokter I, Brommer H, Smit IH, Levato R, van Weeren PR, Castilho M, Malda J. Orthotopic equine study confirms the pivotal importance of structural reinforcement over the pre-culture of cartilage implants. Bioeng Transl Med 2024; 9:e10614. [PMID: 38193127 PMCID: PMC10771555 DOI: 10.1002/btm2.10614] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 01/10/2024] Open
Abstract
In articular cartilage (AC), the collagen arcades provide the tissue with its extraordinary mechanical properties. As these structures cannot be restored once damaged, functional restoration of AC defects remains a major challenge. We report that the use of a converged bioprinted, osteochondral implant, based on a gelatin methacryloyl cartilage phase, reinforced with precisely patterned melt electrowritten polycaprolactone micrometer-scale fibers in a zonal fashion, inspired by native collagen architecture, can provide long-term mechanically stable neo-tissue in an orthotopic large animal model. The design of this novel implant was achieved via state-of-the-art converging of extrusion-based ceramic printing, melt electrowriting, and extrusion-based bioprinting. Interestingly, the cell-free implants, used as a control in this study, showed abundant cell ingrowth and similar favorable results as the cell-containing implants. Our findings underscore the hypothesis that mechanical stability is more determining for the successful survival of the implant than the presence of cells and pre-cultured extracellular matrix. This observation is of great translational importance and highlights the aptness of advanced 3D (bio)fabrication technologies for functional tissue restoration in the harsh articular joint mechanical environment.
Collapse
Affiliation(s)
- Mylène de Ruijter
- Department of Orthopaedics, RMCU Utrecht, UMC UtrechtUniversity of UtrechtUtrechtThe Netherlands
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Paweena Diloksumpan
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Inge Dokter
- Department of Orthopaedics, RMCU Utrecht, UMC UtrechtUniversity of UtrechtUtrechtThe Netherlands
| | - Harold Brommer
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Ineke H. Smit
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, RMCU Utrecht, UMC UtrechtUniversity of UtrechtUtrechtThe Netherlands
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - P. René van Weeren
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Miguel Castilho
- Department of Orthopaedics, RMCU Utrecht, UMC UtrechtUniversity of UtrechtUtrechtThe Netherlands
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
| | - Jos Malda
- Department of Orthopaedics, RMCU Utrecht, UMC UtrechtUniversity of UtrechtUtrechtThe Netherlands
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
46
|
Liang K. Tissue Bioprinting: Promise and Challenges. Bioengineering (Basel) 2023; 10:1400. [PMID: 38135991 PMCID: PMC10740401 DOI: 10.3390/bioengineering10121400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
In recent years, we have witnessed remarkable progress in the field of regenerative medicine, in large part fuelled by developments in advanced biofabrication technologies such as three-dimensional (3D) bioprinting [...].
Collapse
Affiliation(s)
- Kun Liang
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore S138648, Singapore
| |
Collapse
|
47
|
Liu S, Fang C, Zhong C, Li J, Xiao Q. Recent advances in pluripotent stem cell-derived cardiac organoids and heart-on-chip applications for studying anti-cancer drug-induced cardiotoxicity. Cell Biol Toxicol 2023; 39:2527-2549. [PMID: 37889357 DOI: 10.1007/s10565-023-09835-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
Cardiovascular disease (CVD) caused by anti-cancer drug-induced cardiotoxicity is now the second leading cause of mortality among cancer survivors. It is necessary to establish efficient in vitro models for early predicting the potential cardiotoxicity of anti-cancer drugs, as well as for screening drugs that would alleviate cardiotoxicity during and post treatment. Human induced pluripotent stem cells (hiPSCs) have opened up new avenues in cardio-oncology. With the breakthrough of tissue engineering technology, a variety of hiPSC-derived cardiac microtissues or organoids have been recently reported, which have shown enormous potential in studying cardiotoxicity. Moreover, using hiPSC-derived heart-on-chip for studying cardiotoxicity has provided novel insights into the underlying mechanisms. Herein, we summarize different types of anti-cancer drug-induced cardiotoxicities and present an extensive overview on the applications of hiPSC-derived cardiac microtissues, cardiac organoids, and heart-on-chips in cardiotoxicity. Finally, we highlight clinical and translational challenges around hiPSC-derived cardiac microtissues/organoids/heart-on chips and their applications in anti-cancer drug-induced cardiotoxicity. • Anti-cancer drug-induced cardiotoxicities represent pressing challenges for cancer treatments, and cardiovascular disease is the second leading cause of mortality among cancer survivors. • Newly reported in vitro models such as hiPSC-derived cardiac microtissues/organoids/chips show enormous potential for studying cardio-oncology. • Emerging evidence supports that hiPSC-derived cardiac organoids and heart-on-chip are promising in vitro platforms for predicting and minimizing anti-cancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Silin Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London, EC1M 6BQ, UK
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chongkai Fang
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London, EC1M 6BQ, UK
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chong Zhong
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jing Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London, EC1M 6BQ, UK.
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
48
|
Lewns FK, Tsigkou O, Cox LR, Wildman RD, Grover LM, Poologasundarampillai G. Hydrogels and Bioprinting in Bone Tissue Engineering: Creating Artificial Stem-Cell Niches for In Vitro Models. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301670. [PMID: 37087739 PMCID: PMC11478930 DOI: 10.1002/adma.202301670] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Advances in bioprinting have enabled the fabrication of complex tissue constructs with high speed and resolution. However, there remains significant structural and biological complexity within tissues that bioprinting is unable to recapitulate. Bone, for example, has a hierarchical organization ranging from the molecular to whole organ level. Current bioprinting techniques and the materials employed have imposed limits on the scale, speed, and resolution that can be achieved, rendering the technique unable to reproduce the structural hierarchies and cell-matrix interactions that are observed in bone. The shift toward biomimetic approaches in bone tissue engineering, where hydrogels provide biophysical and biochemical cues to encapsulated cells, is a promising approach to enhancing the biological function and development of tissues for in vitro modeling. A major focus in bioprinting of bone tissue for in vitro modeling is creating dynamic microenvironmental niches to support, stimulate, and direct the cellular processes for bone formation and remodeling. Hydrogels are ideal materials for imitating the extracellular matrix since they can be engineered to present various cues whilst allowing bioprinting. Here, recent advances in hydrogels and 3D bioprinting toward creating a microenvironmental niche that is conducive to tissue engineering of in vitro models of bone are reviewed.
Collapse
Affiliation(s)
| | - Olga Tsigkou
- Department of MaterialsUniversity of ManchesterManchesterM1 5GFUK
| | - Liam R. Cox
- School of ChemistryUniversity of BirminghamBirminghamB15 2TTUK
| | - Ricky D. Wildman
- Faculty of EngineeringUniversity of NottinghamNottinghamNG7 2RDUK
| | - Liam M. Grover
- Healthcare Technologies InstituteSchool of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | | |
Collapse
|
49
|
Cianciosi A, Stecher S, Löffler M, Bauer‐Kreisel P, Lim KS, Woodfield TBF, Groll J, Blunk T, Jungst T. Flexible Allyl-Modified Gelatin Photoclick Resin Tailored for Volumetric Bioprinting of Matrices for Soft Tissue Engineering. Adv Healthc Mater 2023; 12:e2300977. [PMID: 37699146 PMCID: PMC11468070 DOI: 10.1002/adhm.202300977] [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: 03/27/2023] [Revised: 08/11/2023] [Indexed: 09/14/2023]
Abstract
Volumetric bioprinting (VBP) is a light-based 3D printing platform, which recently prompted a paradigm shift for additive manufacturing (AM) techniques considering its capability to enable the fabrication of complex cell-laden geometries in tens of seconds with high spatiotemporal control and pattern accuracy. A flexible allyl-modified gelatin (gelAGE)-based photoclick resin is developed in this study to fabricate matrices with exceptionally soft polymer networks (0.2-1.0 kPa). The gelAGE-based resin formulations are designed to exploit the fast thiol-ene crosslinking in combination with a four-arm thiolated polyethylene glycol (PEG4SH) in the presence of a photoinitiator. The flexibility of the gelAGE biomaterial platform allows one to tailor its concentration spanning from 2.75% to 6% and to vary the allyl to thiol ratio without hampering the photocrosslinking efficiency. The thiol-ene crosslinking enables the production of viable cell-material constructs with a high throughput in tens of seconds. The suitability of the gelAGE-based resins is demonstrated by adipogenic differentiation of adipose-derived stromal cells (ASC) after VBP and by the printing of more fragile adipocytes as a proof-of-concept. Taken together, this study introduces a soft photoclick resin which paves the way for volumetric printing applications toward soft tissue engineering.
Collapse
Affiliation(s)
- Alessandro Cianciosi
- Department of Functional Materials in Medicine and DentistryInstitute of Biofabrication and Functional MaterialsUniversity of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)Pleicherwall 297070WürzburgGermany
| | - Sabrina Stecher
- Department of TraumaHandPlastic and Reconstructive SurgeryUniversity Hospital Würzburg97080WürzburgGermany
| | - Maxi Löffler
- Department of Functional Materials in Medicine and DentistryInstitute of Biofabrication and Functional MaterialsUniversity of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)Pleicherwall 297070WürzburgGermany
| | - Petra Bauer‐Kreisel
- Department of TraumaHandPlastic and Reconstructive SurgeryUniversity Hospital Würzburg97080WürzburgGermany
| | - Khoon S. Lim
- School of Medical SciencesUniversity of SydneySydney2006Australia
| | - Tim B. F. Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering and NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Jürgen Groll
- Department of Functional Materials in Medicine and DentistryInstitute of Biofabrication and Functional MaterialsUniversity of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)Pleicherwall 297070WürzburgGermany
| | - Torsten Blunk
- Department of TraumaHandPlastic and Reconstructive SurgeryUniversity Hospital Würzburg97080WürzburgGermany
| | - Tomasz Jungst
- Department of Functional Materials in Medicine and DentistryInstitute of Biofabrication and Functional MaterialsUniversity of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)Pleicherwall 297070WürzburgGermany
| |
Collapse
|
50
|
Esser TU, Anspach A, Muenzebrock KA, Kah D, Schrüfer S, Schenk J, Heinze KG, Schubert DW, Fabry B, Engel FB. Direct 3D-Bioprinting of hiPSC-Derived Cardiomyocytes to Generate Functional Cardiac Tissues. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305911. [PMID: 37655652 DOI: 10.1002/adma.202305911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/18/2023] [Indexed: 09/02/2023]
Abstract
3D-bioprinting is a promising technology to produce human tissues as drug screening tool or for organ repair. However, direct printing of living cells has proven difficult. Here, a method is presented to directly 3D-bioprint human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes embedded in a collagen-hyaluronic acid ink, generating centimeter-sized functional ring- and ventricle-shaped cardiac tissues in an accurate and reproducible manner. The printed tissues contain hiPSC-derived cardiomyocytes with well-organized sarcomeres and exhibit spontaneous and regular contractions, which persist for several months and are able to contract against passive resistance. Importantly, beating frequencies of the printed cardiac tissues can be modulated by pharmacological stimulation. This approach opens up new possibilities for generating complex functional cardiac tissues as models for advanced drug screening or as tissue grafts for organ repair or replacement.
Collapse
Affiliation(s)
- Tilman U Esser
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| | - Annalise Anspach
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| | - Katrin A Muenzebrock
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| | - Delf Kah
- Department of Physics, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Stefan Schrüfer
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Joachim Schenk
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg (JMU), 97080, Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg (JMU), 97080, Würzburg, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Ben Fabry
- Department of Physics, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| |
Collapse
|