1
|
Liu Y, Huang T, Yap NA, Lim K, Ju LA. Harnessing the power of bioprinting for the development of next-generation models of thrombosis. Bioact Mater 2024; 42:328-344. [PMID: 39295733 PMCID: PMC11408160 DOI: 10.1016/j.bioactmat.2024.08.040] [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: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Thrombosis, a leading cause of cardiovascular morbidity and mortality, involves the formation of blood clots within blood vessels. Current animal models and in vitro systems have limitations in recapitulating the complex human vasculature and hemodynamic conditions, limiting the research in understanding the mechanisms of thrombosis. Bioprinting has emerged as a promising approach to construct biomimetic vascular models that closely mimic the structural and mechanical properties of native blood vessels. This review discusses the key considerations for designing bioprinted vascular conduits for thrombosis studies, including the incorporation of key structural, biochemical and mechanical features, the selection of appropriate biomaterials and cell sources, and the challenges and future directions in the field. The advancements in bioprinting techniques, such as multi-material bioprinting and microfluidic integration, have enabled the development of physiologically relevant models of thrombosis. The future of bioprinted models of thrombosis lies in the integration of patient-specific data, real-time monitoring technologies, and advanced microfluidic platforms, paving the way for personalized medicine and targeted interventions. As the field of bioprinting continues to evolve, these advanced vascular models are expected to play an increasingly important role in unraveling the complexities of thrombosis and improving patient outcomes. The continued advancements in bioprinting technologies and the collaboration between researchers from various disciplines hold great promise for revolutionizing the field of thrombosis research.
Collapse
Affiliation(s)
- Yanyan Liu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tao Huang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Nicole Alexis Yap
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Khoon Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- School of Medical Sciences, The University of Sydney, Darlington, NSW 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| |
Collapse
|
2
|
Li W, Li J, Pan C, Lee JS, Kim BS, Gao G. Light-based 3D bioprinting techniques for illuminating the advances of vascular tissue engineering. Mater Today Bio 2024; 29:101286. [PMID: 39435375 PMCID: PMC11492625 DOI: 10.1016/j.mtbio.2024.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Vascular tissue engineering faces significant challenges in creating in vitro vascular disease models, implantable vascular grafts, and vascularized tissue/organ constructs due to limitations in manufacturing precision, structural complexity, replicating the composited architecture, and mimicking the mechanical properties of natural vessels. Light-based 3D bioprinting, leveraging the unique advantages of light including high resolution, rapid curing, multi-material adaptability, and tunable photochemistry, offers transformative solutions to these obstacles. With the emergence of diverse light-based 3D bioprinting techniques and innovative strategies, the advances in vascular tissue engineering have been significantly accelerated. This review provides an overview of the human vascular system and its physiological functions, followed by an in-depth discussion of advancements in light-based 3D bioprinting, including light-dominated and light-assisted techniques. We explore the application of these technologies in vascular tissue engineering for creating in vitro vascular disease models recapitulating key pathological features, implantable blood vessel grafts, and tissue analogs with the integration of capillary-like vasculatures. Finally, we provide readers with insights into the future perspectives of light-based 3D bioprinting to revolutionize vascular tissue engineering.
Collapse
Affiliation(s)
- Wei Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology (BIT), Zhuhai 519088, China
| | - Chen Pan
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, China
- School of Mechanical and Equipment Engineering, Hebei University of Engineering, Handan, 050024, China
| | - Jae-Seong Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
3
|
Dong Y, Zhou X, Ding Y, Luo Y, Zhao H. Advances in tumor microenvironment: Applications and challenges of 3D bioprinting. Biochem Biophys Res Commun 2024; 730:150339. [PMID: 39032359 DOI: 10.1016/j.bbrc.2024.150339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024]
Abstract
The tumor microenvironment (TME) assumes a pivotal role in the treatment of oncological diseases, given its intricate interplay of diverse cellular components and extracellular matrices. This dynamic ecosystem poses a serious challenge to traditional research methods in many ways, such as high research costs, inefficient translation, poor reproducibility, and low modeling success rates. These challenges require the search for more suitable research methods to accurately model the TME, and the emergence of 3D bioprinting technology is transformative and an important complement to these traditional methods to precisely control the distribution of cells, biomolecules, and matrix scaffolds within the TME. Leveraging digital design, the technology enables personalized studies with high precision, providing essential experimental flexibility. Serving as a critical bridge between in vitro and in vivo studies, 3D bioprinting facilitates the realistic 3D culturing of cancer cells. This comprehensive article delves into cutting-edge developments in 3D bioprinting, encompassing diverse methodologies, biomaterial choices, and various 3D tumor models. Exploration of current challenges, including limited biomaterial options, printing accuracy constraints, low reproducibility, and ethical considerations, contributes to a nuanced understanding. Despite these challenges, the technology holds immense potential for simulating tumor tissues, propelling personalized medicine, and constructing high-resolution organ models, marking a transformative trajectory in oncological research.
Collapse
Affiliation(s)
- Yingying Dong
- The First School of Climical Medicine of Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xue Zhou
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Yunyi Ding
- Department of Emergency Medicine, The Second Affiliated Hospital of Zhejiang University, School, Hangzhou, 310009, China.
| | - Yichen Luo
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Hong Zhao
- The First School of Climical Medicine of Zhejiang Chinese Medical University, Hangzhou, 310053, China; Department of Breast Surgery, The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310060, China.
| |
Collapse
|
4
|
Rodrigues FAP, Oliveira CS, Sá SC, Tavaria FK, Lee SJ, Oliveira AL, Costa JB. Molecules in Motion: Unravelling the Dynamics of Vascularization Control in Tissue Engineering. Macromol Biosci 2024:e2400139. [PMID: 39422632 DOI: 10.1002/mabi.202400139] [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: 03/25/2024] [Revised: 08/14/2024] [Indexed: 10/19/2024]
Abstract
Significant progress has been made in tissue engineering (TE), aiming at providing personalized solutions and overcoming the current limitations of traditional tissue and organ transplantation. 3D bioprinting has emerged as a transformative technology in the field, able to mimic key properties of the natural architecture of the native tissues. However, most successes in the area are still limited to avascular or thin tissues due to the difficulties in controlling the vascularization of the engineered tissues. To address this issue, several molecules, biomaterials, and cells with pro- and anti-angiogenic potential have been intensively investigated. Furthermore, different bioreactors capable to provide a dynamic environment for in vitro vascularization control have been also explored. The present review summarizes the main molecules and TE strategies used to promote and inhibit vascularization in TE, as well as the techniques used to deliver them. Additionally, it also discusses the current challenges in 3D bioprinting and in tissue maturation to control in vitro/in vivo vascularization. Currently, this field of investigation is of utmost importance and may open doors for the design and development of more precise and controlled vascularization strategies in TE.
Collapse
Affiliation(s)
- Francisco A P Rodrigues
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Cláudia S Oliveira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Simone C Sá
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Freni K Tavaria
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Ana L Oliveira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - João B Costa
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| |
Collapse
|
5
|
Wheeler EE, Leach JK. Tissue-Engineered Three-Dimensional Platforms for Disease Modeling and Therapeutic Development. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39345164 DOI: 10.1089/ten.teb.2024.0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Three-dimensional (3D) tissue-engineered models are under investigation to recapitulate tissue architecture and functionality, thereby overcoming limitations of traditional two-dimensional cultures and preclinical animal models. This review highlights recent developments in 3D platforms designed to model diseases in vitro that affect numerous tissues and organs, including cardiovascular, gastrointestinal, bone marrow, neural, reproductive, and pulmonary systems. We discuss current technologies for engineered tissue models, highlighting the advantages, limitations, and important considerations for modeling tissues and diseases. Lastly, we discuss future advancements necessary to enhance the reliability of 3D models of tissue development and disease.
Collapse
Affiliation(s)
- Erika E Wheeler
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California, Davis, California, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California, Davis, California, USA
| |
Collapse
|
6
|
Cui X, Jiao J, Yang L, Wang Y, Jiang W, Yu T, Li M, Zhang H, Chao B, Wang Z, Wu M. Advanced tumor organoid bioprinting strategy for oncology research. Mater Today Bio 2024; 28:101198. [PMID: 39205873 PMCID: PMC11357813 DOI: 10.1016/j.mtbio.2024.101198] [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: 04/10/2024] [Revised: 07/14/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Bioprinting is a groundbreaking technology that enables precise distribution of cell-containing bioinks to construct organoid models that accurately reflect the characteristics of tumors in vivo. By incorporating different types of tumor cells into the bioink, the heterogeneity of tumors can be replicated, enabling studies to simulate real-life situations closely. Precise reproduction of the arrangement and interactions of tumor cells using bioprinting methods provides a more realistic representation of the tumor microenvironment. By mimicking the complexity of the tumor microenvironment, the growth patterns and diffusion of tumors can be demonstrated. This approach can also be used to evaluate the response of tumors to drugs, including drug permeability and cytotoxicity, and other characteristics. Therefore, organoid models can provide a more accurate oncology research and treatment simulation platform. This review summarizes the latest advancements in bioprinting to construct tumor organoid models. First, we describe the bioink used for tumor organoid model construction, followed by an introduction to various bioprinting methods for tumor model formation. Subsequently, we provide an overview of existing bioprinted tumor organoid models.
Collapse
Affiliation(s)
- Xiangran Cui
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Jianhang Jiao
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Lili Yang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Yang Wang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Weibo Jiang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Tong Yu
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Mufeng Li
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Han Zhang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Bo Chao
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Zhonghan Wang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - Minfei Wu
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| |
Collapse
|
7
|
Li QQ, Xu D, Dong QW, Song XJ, Chen YB, Cui YL. Biomedical potentials of alginate via physical, chemical, and biological modifications. Int J Biol Macromol 2024; 277:134409. [PMID: 39097042 DOI: 10.1016/j.ijbiomac.2024.134409] [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: 04/12/2024] [Revised: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Alginate is a linear polysaccharide with a modifiable structure and abundant functional groups, offers immense potential for tailoring diverse alginate-based materials to meet the demands of biomedical applications. Given the advancements in modification techniques, it is significant to analyze and summarize the modification of alginate by physical, chemical and biological methods. These approaches provide plentiful information on the preparation, characterization and application of alginate-based materials. Physical modification generally involves blending and physical crosslinking, while chemical modification relies on chemical reactions, mainly including acylation, sulfation, phosphorylation, carbodiimide coupling, nucleophilic substitution, graft copolymerization, terminal modification, and degradation. Chemical modified alginate contains chemically crosslinked alginate, grafted alginate and oligo-alginate. Biological modification associated with various enzymes to realize the hydrolysis or grafting. These diverse modifications hold great promise in fully harnessing the potential of alginate for its burgeoning biomedical applications in the future. In summary, this review provides a comprehensive discussion and summary of different modification methods applied to improve the properties of alginate while expanding its biomedical potentials.
Collapse
Affiliation(s)
- Qiao-Qiao Li
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Dong Xu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Qin-Wei Dong
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Xu-Jiao Song
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Yi-Bing Chen
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
8
|
Wawszczak A, Kocki J, Kołodyńska D. Alginate as a Sustainable and Biodegradable Material for Medical and Environmental Applications-The Case Studies. J Biomed Mater Res B Appl Biomater 2024; 112:1-23. [PMID: 39269132 DOI: 10.1002/jbm.b.35475] [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: 12/13/2023] [Revised: 06/19/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024]
Abstract
Alginates are salts of alginic acid derived mainly from sea algae of the genus brown algae. They are also synthesized by some bacteria. They belong to negatively charged polysaccharides exhibiting some rheological properties. High plasticity and the ability to modify the structure are the reasons for their application in numerous industries. Moreover, when in contact with the living tissue, they do not trigger an immune response, and for this reason they are the most often tested materials for medical applications. The paper discusses the latest applications, including 3D bioprinting, drug delivery systems, and sorptive properties. Recognizing alginates as biomaterials, it emphasizes the necessity for precise processing and modification to industrialize them for specific uses. This review aims to provide a thorough understanding of the advancements in alginate research, underscoring their potential for innovative applications.
Collapse
Affiliation(s)
- Alicja Wawszczak
- Department of Inorganic Chemistry, Faculty of Chemistry, Maria Curie-Skłodowska University, Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Dorota Kołodyńska
- Department of Inorganic Chemistry, Faculty of Chemistry, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
9
|
Stankey PP, Kroll KT, Ainscough AJ, Reynolds DS, Elamine A, Fichtenkort BT, Uzel SGM, Lewis JA. Embedding Biomimetic Vascular Networks via Coaxial Sacrificial Writing into Functional Tissue. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401528. [PMID: 39092638 DOI: 10.1002/adma.202401528] [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: 01/29/2024] [Revised: 07/10/2024] [Indexed: 08/04/2024]
Abstract
Printing human tissues and organs replete with biomimetic vascular networks is of growing interest. While it is possible to embed perfusable channels within acellular and densely cellular matrices, they do not currently possess the biomimetic architectures found in native vessels. Here, coaxial sacrificial writing into functional tissues (co-SWIFT) is developed, an embedded bioprinting method capable of generating hierarchically branching, multilayered vascular networks within both granular hydrogel and densely cellular matrices. Coaxial printheads are designed with an extended core-shell configuration to facilitate robust core-core and shell-shell interconnections between printed branching vessels during embedded bioprinting. Using optimized core-shell ink combinations, biomimetic vessels composed of a smooth muscle cell-laden shell that surrounds perfusable lumens are coaxially printed into granular matrices composed of: 1) transparent alginate microparticles, 2) sacrificial microparticle-laden collagen, or 3) cardiac spheroids derived from human induced pluripotent stem cells. Biomimetic blood vessels that exhibit good barrier function are produced by seeding these interconnected lumens with a confluent layer of endothelial cells. Importantly, it is found that co-SWIFT cardiac tissues mature under perfusion, beat synchronously, and exhibit a cardio-effective drug response in vitro. This advance opens new avenues for the scalable biomanufacturing of vascularized organ-specific tissues for drug testing, disease modeling, and therapeutic use.
Collapse
Affiliation(s)
- Paul P Stankey
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Katharina T Kroll
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Alexander J Ainscough
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Daniel S Reynolds
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Alexander Elamine
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Ben T Fichtenkort
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sebastien G M Uzel
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jennifer A Lewis
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| |
Collapse
|
10
|
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
|
11
|
Nain A, Joshi A, Debnath S, Choudhury S, Thomas J, Satija J, Huang CC, Chatterjee K. A 4D printed nanoengineered super bioactive hydrogel scaffold with programmable deformation for potential bifurcated vascular channel construction. J Mater Chem B 2024; 12:7604-7617. [PMID: 38984474 DOI: 10.1039/d4tb00498a] [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: 07/11/2024]
Abstract
Four-dimensional (4D) printing of hydrogels enabled the fabrication of complex scaffold geometries out of static parts. Although current 4D fabrication strategies are promising for creating vascular parts such as tubes, developing branched networks or tubular junctions is still challenging. Here, for the first time, a 4D printing approach is employed to fabricate T-shaped perfusable bifurcation using an extrusion-based multi-material 3D printing process. An alginate/methylcellulose-based dual-component hydrogel system (with defined swelling behavior) is nanoengineered with carbonized alginate (∼100 nm) to introduce anti-oxidative, anti-inflammatory, and anti-thrombotic properties and shape-shifting properties. A computational model to predict shape deformations in the printed hydrogels with defined infill angles was designed and further validated experimentally. Shape deformations of the 3D-printed flat sheets were achieved by ionic cross-linking. An undisrupted perfusion of a dye solution through a T-junction with minimal leakage mimicking blood flow through vessels is also demonstrated. Moreover, human umbilical vein endothelial and fibroblast cells seeded with printed constructs show intact morphology and excellent cell viability. Overall, the developed strategy paves the way for manufacturing self-actuated vascular bifurcations with remarkable anti-thrombotic properties to potentially treat coronary artery diseases.
Collapse
Affiliation(s)
- Amit Nain
- Department of Material Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India.
- Department of Applied Mechanics & Biomedical Engineering, Indian Institute of Technology-Madras, 600036, Tamil Nadu, India
| | - Akshat Joshi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Souvik Debnath
- Department of Material Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India.
| | - Saswat Choudhury
- Department of Bioengineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Jobin Thomas
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Jitendra Satija
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Chih-Ching Huang
- Department of Bioscience and Biotechnology and Centre of Excellence for the Oceans, National Taiwan Ocean University, Keelung, 202301, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Kaushik Chatterjee
- Department of Material Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| |
Collapse
|
12
|
Wang X, Chen Q, Li J, Tian W, Liu Z, Chen T. Recent adavances of functional modules for tooth regeneration. J Mater Chem B 2024; 12:7497-7518. [PMID: 39021127 DOI: 10.1039/d4tb01027b] [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: 07/20/2024]
Abstract
Dental diseases, such as dental caries and periodontal disorders, constitute a major global health challenge, affecting millions worldwide and often resulting in tooth loss. Traditional dental treatments, though beneficial, typically cannot fully restore the natural functions and structures of teeth. This limitation has prompted growing interest in innovative strategies for tooth regeneration methods. Among these, the use of dental stem cells to generate functional tooth modules represents an emerging and promising approach in dental tissue engineering. These modules aim to closely replicate the intricate morphology and essential physiological functions of dental tissues. Recent advancements in regenerative research have not only enhanced the assembly techniques for these modules but also highlighted their therapeutic potential in addressing various dental diseases. In this review, we discuss the latest progress in the construction of functional tooth modules, especially on regenerating dental pulp, periodontal tissue, and tooth roots.
Collapse
Affiliation(s)
- Xuan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Qiuyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jiayi Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhi Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Tian Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
13
|
Zhao N, Pessell AF, Zhu N, Searson PC. Tissue-Engineered Microvessels: A Review of Current Engineering Strategies and Applications. Adv Healthc Mater 2024; 13:e2303419. [PMID: 38686434 PMCID: PMC11338730 DOI: 10.1002/adhm.202303419] [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: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Microvessels, including arterioles, capillaries, and venules, play an important role in regulating blood flow, enabling nutrient and waste exchange, and facilitating immune surveillance. Due to their important roles in maintaining normal function in human tissues, a substantial effort has been devoted to developing tissue-engineered models to study endothelium-related biology and pathology. Various engineering strategies have been developed to recapitulate the structural, cellular, and molecular hallmarks of native human microvessels in vitro. In this review, recent progress in engineering approaches, key components, and culture platforms for tissue-engineered human microvessel models is summarized. Then, tissue-specific models, and the major applications of tissue-engineered microvessels in development, disease modeling, drug screening and delivery, and vascularization in tissue engineering, are reviewed. Finally, future research directions for the field are discussed.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Alexander F Pessell
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ninghao Zhu
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
14
|
Fang W, Yu Z, Gao G, Yang M, Du X, Wang Y, Fu Q. Light-based 3D bioprinting technology applied to repair and regeneration of different tissues: A rational proposal for biomedical applications. Mater Today Bio 2024; 27:101135. [PMID: 39040222 PMCID: PMC11262185 DOI: 10.1016/j.mtbio.2024.101135] [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: 02/02/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
3D bioprinting technology, a subset of 3D printing technology, is currently witnessing widespread utilization in tissue repair and regeneration endeavors. In particular, light-based 3D bioprinting technology has garnered significant interest and favor. Central to its successful implementation lies the judicious selection of photosensitive polymers. Moreover, by fine-tuning parameters such as light irradiation time, choice of photoinitiators and crosslinkers, and their concentrations, the properties of the scaffolds can be tailored to suit the specific requirements of the targeted tissue repair sites. In this comprehensive review, we provide an overview of commonly utilized bio-inks suitable for light-based 3D bioprinting, delving into the distinctive characteristics of each material. Furthermore, we delineate strategies for bio-ink selection tailored to diverse repair locations, alongside methods for optimizing printing parameters. Ultimately, we present a coherent synthesis aimed at enhancing the practical application of light-based 3D bioprinting technology in tissue engineering, while also addressing current challenges and future prospects.
Collapse
Affiliation(s)
- Wenzhuo Fang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Zhenwei Yu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Guo Gao
- Key Laboratory for Thin Film and Micro Fabrication of the Ministry of Education, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ming Yang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Xuan Du
- Key Laboratory for Thin Film and Micro Fabrication of the Ministry of Education, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ying Wang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Qiang Fu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| |
Collapse
|
15
|
Seow KS, Ling APK. Mesenchymal stem cells as future treatment for cardiovascular regeneration and its challenges. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:73. [PMID: 39118948 PMCID: PMC11304428 DOI: 10.21037/atm-23-1936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 12/04/2023] [Indexed: 08/10/2024]
Abstract
Cardiovascular diseases (CVDs), particularly stroke and myocardial infarction (MI) contributed to the leading cause of death annually among the chronic diseases globally. Despite the advancement of technology, the current available treatments mainly served as palliative care but not treating the diseases. However, the discovery of mesenchymal stem cells (MSCs) had gained a consideration to serve as promising strategy in treating CVDs. Recent evidence also showed that MSCs are the strong candidate to be used as stem cell therapy involving cardiovascular regeneration due to its cardiomyogenesis, anti-inflammatory and immunomodulatory properties, antifibrotic effects and neovascularization capacity. Besides, MSCs could be used for cellular cardiomyoplasty with its transdifferentiation of MSCs into cardiomyocytes, paracrine effects, microvesicles and exosomes as well as mitochondrial transfer. The safety and efficacy of utilizing MSCs have been described in well-established preclinical and clinical studies in which the accomplishment of MSCs transplantation resulted in further improvement of the cardiac function. Tissue engineering could enhance the desired properties and therapeutic effects of MSCs in cardiovascular regeneration by genome-editing, facilitating the cell delivery and retention, biomaterials-based scaffold, and three-dimensional (3D)-bioprinting. However, there are still obstacles in the use of MSCs due to the complexity and versatility of MSCs, low retention rate, route of administration and the ethical and safety issues of the use of MSCs. The aim of this review is to highlight the details of therapeutic properties of MSCs in treating CVDs, strategies to facilitate the therapeutic effects of MSCs through tissue engineering and the challenges faced using MSCs. A comprehensive review has been done through PubMed and National Center for Biotechnology Information (NCBI) from the year of 2010 to 2021 based on some specific key terms such as 'mesenchymal stem cells in cardiovascular disease', 'mesenchymal stem cells in cardiac regeneration', 'mesenchymal stem cells facilitate cardiac repairs', 'tissue engineering of MSCs' to include relevant literature in this review.
Collapse
Affiliation(s)
- Ke Sin Seow
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Anna Pick Kiong Ling
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Martier A, Chen Z, Schaps H, Mondrinos MJ, Fang JS. Capturing physiological hemodynamic flow and mechanosensitive cell signaling in vessel-on-a-chip platforms. Front Physiol 2024; 15:1425618. [PMID: 39135710 PMCID: PMC11317428 DOI: 10.3389/fphys.2024.1425618] [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: 05/06/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Recent advances in organ chip (or, "organ-on-a-chip") technologies and microphysiological systems (MPS) have enabled in vitro investigation of endothelial cell function in biomimetic three-dimensional environments under controlled fluid flow conditions. Many current organ chip models include a vascular compartment; however, the design and implementation of these vessel-on-a-chip components varies, with consequently varied impact on their ability to capture and reproduce hemodynamic flow and associated mechanosensitive signaling that regulates key characteristics of healthy, intact vasculature. In this review, we introduce organ chip and vessel-on-a-chip technology in the context of existing in vitro and in vivo vascular models. We then briefly discuss the importance of mechanosensitive signaling for vascular development and function, with focus on the major mechanosensitive signaling pathways involved. Next, we summarize recent advances in MPS and organ chips with an integrated vascular component, with an emphasis on comparing both the biomimicry and adaptability of the diverse approaches used for supporting and integrating intravascular flow. We review current data showing how intravascular flow and fluid shear stress impacts vessel development and function in MPS platforms and relate this to existing work in cell culture and animal models. Lastly, we highlight new insights obtained from MPS and organ chip models of mechanosensitive signaling in endothelial cells, and how this contributes to a deeper understanding of vessel growth and function in vivo. We expect this review will be of broad interest to vascular biologists, physiologists, and cardiovascular physicians as an introduction to organ chip platforms that can serve as viable model systems for investigating mechanosensitive signaling and other aspects of vascular physiology.
Collapse
Affiliation(s)
- A. Martier
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - Z. Chen
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - H. Schaps
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - M. J. Mondrinos
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States
| | - J. S. Fang
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States
| |
Collapse
|
17
|
Salih T, Caputo M, Ghorbel MT. Recent Advances in Hydrogel-Based 3D Bioprinting and Its Potential Application in the Treatment of Congenital Heart Disease. Biomolecules 2024; 14:861. [PMID: 39062575 PMCID: PMC11274841 DOI: 10.3390/biom14070861] [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: 05/15/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Congenital heart disease (CHD) is the most common birth defect, requiring invasive surgery often before a child's first birthday. Current materials used during CHD surgery lack the ability to grow, remodel, and regenerate. To solve those limitations, 3D bioprinting is an emerging tool with the capability to create tailored constructs based on patients' own imaging data with the ability to grow and remodel once implanted in children with CHD. It has the potential to integrate multiple bioinks with several cell types and biomolecules within 3D-bioprinted constructs that exhibit good structural fidelity, stability, and mechanical integrity. This review gives an overview of CHD and recent advancements in 3D bioprinting technologies with potential use in the treatment of CHD. Moreover, the selection of appropriate biomaterials based on their chemical, physical, and biological properties that are further manipulated to suit their application are also discussed. An introduction to bioink formulations composed of various biomaterials with emphasis on multiple cell types and biomolecules is briefly overviewed. Vasculogenesis and angiogenesis of prefabricated 3D-bioprinted structures and novel 4D printing technology are also summarized. Finally, we discuss several restrictions and our perspective on future directions in 3D bioprinting technologies in the treatment of CHD.
Collapse
Affiliation(s)
- Tasneem Salih
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK; (T.S.); (M.C.)
| | - Massimo Caputo
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK; (T.S.); (M.C.)
- Cardiac Surgery, University Hospitals Bristol, NHS Foundation Trust, Bristol BS2 8HW, UK
| | - Mohamed T. Ghorbel
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK; (T.S.); (M.C.)
| |
Collapse
|
18
|
Khodadadi Yazdi M, Seidi F, Hejna A, Zarrintaj P, Rabiee N, Kucinska-Lipka J, Saeb MR, Bencherif SA. Tailor-Made Polysaccharides for Biomedical Applications. ACS APPLIED BIO MATERIALS 2024; 7:4193-4230. [PMID: 38958361 PMCID: PMC11253104 DOI: 10.1021/acsabm.3c01199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
Polysaccharides (PSAs) are carbohydrate-based macromolecules widely used in the biomedical field, either in their pure form or in blends/nanocomposites with other materials. The relationship between structure, properties, and functions has inspired scientists to design multifunctional PSAs for various biomedical applications by incorporating unique molecular structures and targeted bulk properties. Multiple strategies, such as conjugation, grafting, cross-linking, and functionalization, have been explored to control their mechanical properties, electrical conductivity, hydrophilicity, degradability, rheological features, and stimuli-responsiveness. For instance, custom-made PSAs are known for their worldwide biomedical applications in tissue engineering, drug/gene delivery, and regenerative medicine. Furthermore, the remarkable advancements in supramolecular engineering and chemistry have paved the way for mission-oriented biomaterial synthesis and the fabrication of customized biomaterials. These materials can synergistically combine the benefits of biology and chemistry to tackle important biomedical questions. Herein, we categorize and summarize PSAs based on their synthesis methods, and explore the main strategies used to customize their chemical structures. We then highlight various properties of PSAs using practical examples. Lastly, we thoroughly describe the biomedical applications of tailor-made PSAs, along with their current existing challenges and potential future directions.
Collapse
Affiliation(s)
- Mohsen Khodadadi Yazdi
- Division
of Electrochemistry and Surface Physical Chemistry, Faculty of Applied
Physics and Mathematics, Gdańsk University
of Technology, Narutowicza
11/12, 80-233 Gdańsk, Poland
- Advanced
Materials Center, Gdańsk University
of Technology, Narutowicza
11/12, 80-233 Gdańsk, Poland
| | - Farzad Seidi
- Jiangsu
Co−Innovation Center for Efficient Processing and Utilization
of Forest Resources and International Innovation Center for Forest
Chemicals and Materials, Nanjing Forestry
University, Nanjing 210037, China
| | - Aleksander Hejna
- Institute
of Materials Technology, Poznan University
of Technology, PL-61-138 Poznań, Poland
| | - Payam Zarrintaj
- School
of Chemical Engineering, Oklahoma State
University, 420 Engineering
North, Stillwater, Oklahoma 74078, United States
| | - Navid Rabiee
- Department
of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| | - Justyna Kucinska-Lipka
- Department
of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, 80-233 Gdańsk, Poland
| | - Mohammad Reza Saeb
- Department
of Pharmaceutical Chemistry, Medical University
of Gdańsk, J.
Hallera 107, 80-416 Gdańsk, Poland
| | - Sidi A. Bencherif
- Chemical
Engineering Department, Northeastern University, Boston, Massachusetts 02115, United States
- Department
of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
- Harvard
John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
19
|
Xu C, Alameri A, Leong W, Johnson E, Chen Z, Xu B, Leong KW. Multiscale engineering of brain organoids for disease modeling. Adv Drug Deliv Rev 2024; 210:115344. [PMID: 38810702 PMCID: PMC11265575 DOI: 10.1016/j.addr.2024.115344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.
Collapse
Affiliation(s)
- Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alia Alameri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Emily Johnson
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
20
|
Zhang J, Suttapreyasri S, Leethanakul C, Samruajbenjakun B. Fabrication of vascularized tissue-engineered bone models using triaxial bioprinting. J Biomed Mater Res A 2024; 112:1093-1106. [PMID: 38411369 DOI: 10.1002/jbm.a.37694] [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: 09/15/2023] [Revised: 01/01/2024] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
Bone tissue is a highly vascularized tissue. When constructing tissue-engineered bone models, both the osteogenic and angiogenic capabilities of the construct should be carefully considered. However, fabricating a vascularized tissue-engineered bone to promote vascular formation and bone generation, while simultaneously establishing nutrition channels to facilitate nutrient exchange within the constructs, remains a significant challenge. Triaxial bioprinting, which not only allows the independent encapsulation of different cell types while simultaneously forming nutrient channels, could potentially emerge as a strategy for fabricating vascularized tissue-engineered bone. Moreover, bioinks should also be applied in combination to promote both osteogenesis and angiogenesis. In this study, employing triaxial bioprinting, we used a blend bioink of gelatin methacryloyl (GelMA), sodium alginate (Alg), and different concentrations of nano beta-tricalcium phosphate (nano β-TCP) encapsulated MC3T3-E1 preosteoblasts as the outer layer, a mixed bioink of GelMA and Alg loaded with human umbilical vein endothelial cells (HUVEC) as the middle layer, and gelatin as a sacrificial material to form nutrient channels in the inner layer to fabricate vascularized bone constructs simulating the microenvironment for bone and vascular tissues. The results showed that the addition of nano β-TCP could adjust the mechanical, swelling, and degradation properties of the constructs. Biological assessments revealed the cell viability of constructs containing different concentrations of nano β-TCP was higher than 90% on day 7, The cell-laden constructs containing 3% (w/v) nano β-TCP exhibited better osteogenic (higher Alkaline phosphatase activity and larger Osteocalcin positive area) and angiogenic (the gradual increased CD31 positive area) potential. Therefore, using triaxial bioprinting technology and employing GelMA, Alg, and nano β-TCP as bioink components could fabricate vascularized bone tissue constructs, offering a novel strategy for vascularized bone tissue engineering.
Collapse
Affiliation(s)
- Junbiao Zhang
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
- Guiyang Hospital of Stomatology, Guiyang, People's Republic of China
| | - Srisurang Suttapreyasri
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Thailand
| | - Chidchanok Leethanakul
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
| | - Bancha Samruajbenjakun
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
21
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
22
|
Rössler L, Nasehi R, Hansen N, Aveic S, Fischer H. Porous collagen scaffolds enable endothelial lumen formation in vitro under both static and dynamic growth conditions. J Biomed Mater Res B Appl Biomater 2024; 112:e35444. [PMID: 38923270 DOI: 10.1002/jbm.b.35444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/26/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Despite recent advances in the field of tissue engineering, the development of complex tissue-like structures in vitro is compromised by the lack of integration of a functioning vasculature. In this study, we propose a mesoscale three-dimensional (3D) in vitro vascularized connective tissue model and demonstrate its feasibility to prompt the self-assembly of endothelial cells into vessel-like structures. Moreover, we investigate the effect of perfusion on the organization of the cells. For this purpose, primary endothelial cells (HUVECs) and a cell line of human foreskin fibroblasts are cultivated in ECM-like matrices made up of freeze-dried collagen scaffolds permeated with collagen type I hydrogel. A tailored bioreactor is designed to investigate the effect of perfusion on self-organization of HUVECs. Immunofluorescent staining, two-photon microscopy, second-harmonic generation imaging, and scanning electron microscopy are applied to visualize the spatial arrangement of the cells. The analyses reveal the formation of hollow, vessel-like structures of HUVECs in hydrogel-permeated collagen scaffolds under both static and dynamic conditions. In conclusion, we demonstrate the feasibility of a 3D porous collagen scaffolding system that enables and maintains the self-organization of HUVECs into vessel-like structures independent of a dynamic flow.
Collapse
Affiliation(s)
- Lena Rössler
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
- Department of Preventive Dentistry, Periodontology and Cariology, University Medical Center, Göttingen, Germany
| | - Ramin Nasehi
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
- DWI - Leibniz Institute for Interactive Materials, Aachen, Germany
| | - Nadja Hansen
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Sanja Aveic
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
23
|
Li YB, Rukhlova M, Zhang D, Nhan J, Sodja C, Bedford E, St-Pierre JP, Jezierski A. Single-Step 3D Bioprinting of Alginate-Collagen Type I Hydrogel Fiber Rings to Promote Angiogenic Network Formation. Tissue Eng Part C Methods 2024; 30:289-306. [PMID: 38946589 DOI: 10.1089/ten.tec.2024.0083] [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: 07/02/2024] Open
Abstract
In the advent of tissue engineering and regenerative medicine, the demand for innovative approaches to biofabricate complex vascular structures is increasing. We describe a single-step 3D bioprinting method leveraging Aspect Biosystems RX1 technology, which integrates the crosslinking step at a flow-focusing junction, to biofabricate immortalized adult rat brain endothelial cell (SV-ARBEC)-encapsulated alginate-collagen type I hydrogel rings. This single-step biofabrication process involves the strategic layer-by-layer assembly of hydrogel rings, encapsulating SV-ARBECs in a spatially controlled manner while optimizing access to media and nutrients. The spatial arrangement of the SV-ARBECs within the rings promotes spontaneous angiogenic network formation and the constrained deposition of cells within the hydrogel matrix facilitates tissue-like organized vascular-like network development. This approach provides a platform that can be adapted to many different endothelial cell types and leveraged to better understand the mechanisms driving angiogenesis and vascular-network formation in 3D bioprinted constructs supporting the development of more complex tissue and disease models for advancing drug discovery, tissue engineering, and regenerative medicine applications.
Collapse
Affiliation(s)
- Ying Betty Li
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, Canada
- Department of Systems and Computer Engineering, Faculty of Engineering and Design, Carleton University, Ottawa, Canada
| | - Marina Rukhlova
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, Canada
| | - Dongling Zhang
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, Canada
| | - Jordan Nhan
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada
| | - Caroline Sodja
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, Canada
| | | | - Jean-Philippe St-Pierre
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada
| | - Anna Jezierski
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, Canada
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada
| |
Collapse
|
24
|
Rosellini E, Giordano C, Guidi L, Cascone MG. Biomimetic Approaches in Scaffold-Based Blood Vessel Tissue Engineering. Biomimetics (Basel) 2024; 9:377. [PMID: 39056818 PMCID: PMC11274842 DOI: 10.3390/biomimetics9070377] [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: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Cardiovascular diseases remain a leading cause of mortality globally, with atherosclerosis representing a significant pathological means, often leading to myocardial infarction. Coronary artery bypass surgery, a common procedure used to treat coronary artery disease, presents challenges due to the limited autologous tissue availability or the shortcomings of synthetic grafts. Consequently, there is a growing interest in tissue engineering approaches to develop vascular substitutes. This review offers an updated picture of the state of the art in vascular tissue engineering, emphasising the design of scaffolds and dynamic culture conditions following a biomimetic approach. By emulating native vessel properties and, in particular, by mimicking the three-layer structure of the vascular wall, tissue-engineered grafts can improve long-term patency and clinical outcomes. Furthermore, ongoing research focuses on enhancing biomimicry through innovative scaffold materials, surface functionalisation strategies, and the use of bioreactors mimicking the physiological microenvironment. Through a multidisciplinary lens, this review provides insight into the latest advancements and future directions of vascular tissue engineering, with particular reference to employing biomimicry to create systems capable of reproducing the structure-function relationships present in the arterial wall. Despite the existence of a gap between benchtop innovation and clinical translation, it appears that the biomimetic technologies developed to date demonstrate promising results in preventing vascular occlusion due to blood clotting under laboratory conditions and in preclinical studies. Therefore, a multifaceted biomimetic approach could represent a winning strategy to ensure the translation of vascular tissue engineering into clinical practice.
Collapse
Affiliation(s)
- Elisabetta Rosellini
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy; (C.G.); (L.G.)
| | | | | | - Maria Grazia Cascone
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy; (C.G.); (L.G.)
| |
Collapse
|
25
|
Simińska-Stanny J, Nicolas L, Chafai A, Jafari H, Hajiabbas M, Dodi G, Gardikiotis I, Delporte C, Nie L, Podstawczyk D, Shavandi A. Advanced PEG-tyramine biomaterial ink for precision engineering of perfusable and flexible small-diameter vascular constructs via coaxial printing. Bioact Mater 2024; 36:168-184. [PMID: 38463551 PMCID: PMC10924180 DOI: 10.1016/j.bioactmat.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 03/12/2024] Open
Abstract
Vascularization is crucial for providing nutrients and oxygen to cells while removing waste. Despite advances in 3D-bioprinting, the fabrication of structures with void spaces and channels remains challenging. This study presents a novel approach to create robust yet flexible and permeable small (600-1300 μm) artificial vessels in a single processing step using 3D coaxial extrusion printing of a biomaterial ink, based on tyramine-modified polyethylene glycol (PEG-Tyr). We combined the gelatin biocompatibility/activity, robustness of PEG-Tyr and alginate with the shear-thinning properties of methylcellulose (MC) in a new biomaterial ink for the fabrication of bioinspired vessels. Chemical characterization using NMR and FTIR spectroscopy confirmed the successful modification of PEG with Tyr and rheological characterization indicated that the addition of PEG-Tyr decreased the viscosity of the ink. Enzyme-mediated crosslinking of PEG-Tyr allowed the formation of covalent crosslinks within the hydrogel chains, ensuring its stability. PEG-Tyr units improved the mechanical properties of the material, resulting in stretchable and elastic constructs without compromising cell viability and adhesion. The printed vessel structures displayed uniform wall thickness, shape retention, improved elasticity, permeability, and colonization by endothelial-derived - EA.hy926 cells. The chorioallantoic membrane (CAM) and in vivo assays demonstrated the hydrogel's ability to support neoangiogenesis. The hydrogel material with PEG-Tyr modification holds promise for vascular tissue engineering applications, providing a flexible, biocompatible, and functional platform for the fabrication of vascular structures.
Collapse
Affiliation(s)
- Julia Simińska-Stanny
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Lise Nicolas
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- European School of Materials Science and Engineering, University of Lorraine, Nancy, France
| | - Adam Chafai
- Université Libre de Bruxelles (ULB), Micro-milli Platform, Avenue F.D. Roosevelt, 50 - CP 165/67, 1050, Brussels, Belgium
| | - Hafez Jafari
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Maryam Hajiabbas
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- Université Libre de Bruxelles (ULB), Faculté de Médecine, Campus Erasme - CP 611, Laboratory of Pathophysiological and Nutritional Biochemistry, Route de Lennik, 808, 1070, Bruxelles, Belgium
| | - Gianina Dodi
- Faculty of Medical Bioengineering, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Ioannis Gardikiotis
- Advanced Research and Development Center for Experimental Medicine, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Christine Delporte
- Université Libre de Bruxelles (ULB), Faculté de Médecine, Campus Erasme - CP 611, Laboratory of Pathophysiological and Nutritional Biochemistry, Route de Lennik, 808, 1070, Bruxelles, Belgium
| | - Lei Nie
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Daria Podstawczyk
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373, Wroclaw, Poland
| | - Amin Shavandi
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| |
Collapse
|
26
|
Kiratitanaporn W, Guan J, Berry DB, Lao A, Chen S. Multimodal Three-Dimensional Printing for Micro-Modulation of Scaffold Stiffness Through Machine Learning. Tissue Eng Part A 2024; 30:280-292. [PMID: 37747804 DOI: 10.1089/ten.tea.2023.0193] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
The ability to precisely control a scaffold's microstructure and geometry with light-based three-dimensional (3D) printing has been widely demonstrated. However, the modulation of scaffold's mechanical properties through prescribed printing parameters is still underexplored. This study demonstrates a novel 3D-printing workflow to create a complex, elastomeric scaffold with precision-engineered stiffness control by utilizing machine learning. Various printing parameters, including the exposure time, light intensity, printing infill, laser pump current, and printing speed were modulated to print poly (glycerol sebacate) acrylate (PGSA) scaffolds with mechanical properties ranging from 49.3 ± 3.3 kPa to 2.8 ± 0.3 MPa. This enables flexibility in spatial stiffness modulation in addition to high-resolution scaffold fabrication. Then, a neural network-based machine learning model was developed and validated to optimize printing parameters to yield scaffolds with user-defined stiffness modulation for two different vat photopolymerization methods: a digital light processing (DLP)-based 3D printer was utilized to rapidly fabricate stiffness-modulated scaffolds with features on the hundreds of micron scale and a two-photon polymerization (2PP) 3D printer was utilized to print fine structures on the submicron scale. A novel 3D-printing workflow was designed to utilize both DLP-based and 2PP 3D printers to create multiscale scaffolds with precision-tuned stiffness control over both gross and fine geometric features. The described workflow can be used to fabricate scaffolds for a variety of tissue engineering applications, specifically for interfacial tissue engineering for which adjacent tissues possess heterogeneous mechanical properties (e.g., muscle-tendon).
Collapse
Affiliation(s)
- Wisarut Kiratitanaporn
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Jiaao Guan
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, California, USA
| | - David B Berry
- Department of Orthopedic Surgery, University of California San Diego, La Jolla, California, USA
| | - Alison Lao
- Department of NanoEngineering, University of California San Diego, La Jolla, California, USA
| | - Shaochen Chen
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, California, USA
- Department of NanoEngineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
27
|
Kunwar P, Aryal U, Poudel A, Fougnier D, Geffert ZJ, Xie R, Li Z, Soman P. Droplet bioprinting of acellular and cell-laden structures at high-resolutions. Biofabrication 2024; 16:035019. [PMID: 38749419 DOI: 10.1088/1758-5090/ad4c09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/15/2024] [Indexed: 05/24/2024]
Abstract
Advances in digital light projection(DLP) based (bio) printers have made printing of intricate structures at high resolution possible using a wide range of photosensitive bioinks. A typical setup of a DLP bioprinter includes a vat or reservoir filled with liquid bioink, which presents challenges in terms of cost associated with bioink synthesis, high waste, and gravity-induced cell settling, contaminations, or variation in bioink viscosity during the printing process. Here, we report a vat-free, low-volume, waste-free droplet bioprinting method capable of rapidly printing 3D soft structures at high resolution using model bioinks and model cells. A multiphase many-body dissipative particle dynamics model was developed to simulate the dynamic process of droplet-based DLP printing and elucidate the roles of surface wettability and bioink viscosity. Process variables such as light intensity, photo-initiator concentration, and bioink formulations were optimized to print 3D soft structures (∼0.4-3 kPa) with a typical layer thickness of 50µm, an XY resolution of 38 ± 1.5μm and Z resolution of 237 ± 5.4µm. To demonstrate its versatility, droplet bioprinting was used to print a range of acellular 3D structures such as a lattice cube, a Mayan pyramid, a heart-shaped structure, and a microfluidic chip with endothelialized channels. Droplet bioprinting, performed using model C3H/10T1/2 cells, exhibited high viability (90%) and cell spreading. Additionally, microfluidic devices with internal channel networks lined with endothelial cells showed robust monolayer formation while osteoblast-laden constructs showed mineral deposition upon osteogenic induction. Overall, droplet bioprinting could be a low-cost, no-waste, easy-to-use, method to make customized bioprinted constructs for a range of biomedical applications.
Collapse
Affiliation(s)
- Puskal Kunwar
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Ujjwal Aryal
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Arun Poudel
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Daniel Fougnier
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Zachary J Geffert
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Rui Xie
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Zhen Li
- Department of Mechanical Engineering, Clemson University, Clemson, SC 29634, United States of America
| | - Pranav Soman
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| |
Collapse
|
28
|
Shukla AK, Yoon S, Oh SO, Lee D, Ahn M, Kim BS. Advancement in Cancer Vasculogenesis Modeling through 3D Bioprinting Technology. Biomimetics (Basel) 2024; 9:306. [PMID: 38786516 PMCID: PMC11118135 DOI: 10.3390/biomimetics9050306] [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: 04/09/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Cancer vasculogenesis is a pivotal focus of cancer research and treatment given its critical role in tumor development, metastasis, and the formation of vasculogenic microenvironments. Traditional approaches to investigating cancer vasculogenesis face significant challenges in accurately modeling intricate microenvironments. Recent advancements in three-dimensional (3D) bioprinting technology present promising solutions to these challenges. This review provides an overview of cancer vasculogenesis and underscores the importance of precise modeling. It juxtaposes traditional techniques with 3D bioprinting technologies, elucidating the advantages of the latter in developing cancer vasculogenesis models. Furthermore, it explores applications in pathological investigations, preclinical medication screening for personalized treatment and cancer diagnostics, and envisages future prospects for 3D bioprinted cancer vasculogenesis models. Despite notable advancements, current 3D bioprinting techniques for cancer vasculogenesis modeling have several limitations. Nonetheless, by overcoming these challenges and with technological advances, 3D bioprinting exhibits immense potential for revolutionizing the understanding of cancer vasculogenesis and augmenting treatment modalities.
Collapse
Affiliation(s)
- Arvind Kumar Shukla
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sik Yoon
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 50612, Republic of Korea
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 50612, Republic of Korea
| | - Sae-Ock Oh
- Research Center for Molecular Control of Cancer Cell Diversity, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University College of Medicine, Yangsan 50612, Republic of Korea
| | - Minjun Ahn
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
29
|
Raoufinia R, Rahimi HR, Saburi E, Moghbeli M. Advances and challenges of the cell-based therapies among diabetic patients. J Transl Med 2024; 22:435. [PMID: 38720379 PMCID: PMC11077715 DOI: 10.1186/s12967-024-05226-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
Diabetes mellitus is a significant global public health challenge, with a rising prevalence and associated morbidity and mortality. Cell therapy has evolved over time and holds great potential in diabetes treatment. In the present review, we discussed the recent progresses in cell-based therapies for diabetes that provides an overview of islet and stem cell transplantation technologies used in clinical settings, highlighting their strengths and limitations. We also discussed immunomodulatory strategies employed in cell therapies. Therefore, this review highlights key progresses that pave the way to design transformative treatments to improve the life quality among diabetic patients.
Collapse
Affiliation(s)
- Ramin Raoufinia
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Donzanti MJ, Mhatre O, Chernokal B, Renteria DC, Gleghorn JP. Stochastic to Deterministic: A straightforward approach to create serially perfusable multiscale capillary beds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592474. [PMID: 38766003 PMCID: PMC11100595 DOI: 10.1101/2024.05.03.592474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Generation of in vitro tissue models with serially perfused hierarchical vasculature would allow greater control of fluid perfusion throughout the network and enable direct mechanistic investigation of vasculogenesis, angiogenesis, and vascular remodeling. In this work, we have developed a method to produce a closed, serially perfused, multiscale vessel network embedded within an acellular hydrogel. We confirmed that the acellular and cellular gel-gel interface was functionally annealed without preventing or biasing cell migration and endothelial self-assembly. Multiscale connectivity of the vessel network was validated via high-resolution microscopy techniques to confirm anastomosis between self-assembled and patterned vessels. Lastly, using fluorescently labeled microspheres, the multiscale network was serially perfused to confirm patency and barrier function. Directional flow from inlet to outlet man-dated flow through the capillary bed. This method for producing closed, multiscale vascular networks was developed with the intention of straightforward fabrication and engineering techniques so as to be a low barrier to entry for researchers who wish to investigate mechanistic questions in vascular biology. This ease of use offers a facile extension of these methods for incorporation into organoid culture, organ-on-a-chip (OOC) models, and bioprinted tissues.
Collapse
|
31
|
Maharjan S, Ma C, Singh B, Kang H, Orive G, Yao J, Shrike Zhang Y. Advanced 3D imaging and organoid bioprinting for biomedical research and therapeutic applications. Adv Drug Deliv Rev 2024; 208:115237. [PMID: 38447931 PMCID: PMC11031334 DOI: 10.1016/j.addr.2024.115237] [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: 11/08/2023] [Revised: 01/15/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoid cultures offer a valuable platform for studying organ-level biology, allowing for a closer mimicry of human physiology compared to traditional two-dimensional cell culture systems or non-primate animal models. While many organoid cultures use cell aggregates or decellularized extracellular matrices as scaffolds, they often lack precise biochemical and biophysical microenvironments. In contrast, three-dimensional (3D) bioprinting allows precise placement of organoids or spheroids, providing enhanced spatial control and facilitating the direct fusion for the formation of large-scale functional tissues in vitro. In addition, 3D bioprinting enables fine tuning of biochemical and biophysical cues to support organoid development and maturation. With advances in the organoid technology and its potential applications across diverse research fields such as cell biology, developmental biology, disease pathology, precision medicine, drug toxicology, and tissue engineering, organoid imaging has become a crucial aspect of physiological and pathological studies. This review highlights the recent advancements in imaging technologies that have significantly contributed to organoid research. Additionally, we discuss various bioprinting techniques, emphasizing their applications in organoid bioprinting. Integrating 3D imaging tools into a bioprinting platform allows real-time visualization while facilitating quality control, optimization, and comprehensive bioprinting assessment. Similarly, combining imaging technologies with organoid bioprinting can provide valuable insights into tissue formation, maturation, functions, and therapeutic responses. This approach not only improves the reproducibility of physiologically relevant tissues but also enhances understanding of complex biological processes. Thus, careful selection of bioprinting modalities, coupled with appropriate imaging techniques, holds the potential to create a versatile platform capable of addressing existing challenges and harnessing opportunities in these rapidly evolving fields.
Collapse
Affiliation(s)
- Sushila Maharjan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Chenshuo Ma
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Bibhor Singh
- Winthrop L. Chenery Upper Elementary School, Belmont, MA 02478, USA
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea; College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, 01007, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| |
Collapse
|
32
|
Wei Q, An Y, Zhao X, Li M, Zhang J. Three-dimensional bioprinting of tissue-engineered skin: Biomaterials, fabrication techniques, challenging difficulties, and future directions: A review. Int J Biol Macromol 2024; 266:131281. [PMID: 38641503 DOI: 10.1016/j.ijbiomac.2024.131281] [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: 12/31/2023] [Revised: 03/17/2024] [Accepted: 03/29/2024] [Indexed: 04/21/2024]
Abstract
As an emerging new manufacturing technology, Three-dimensional (3D) bioprinting provides the potential for the biomimetic construction of multifaceted and intricate architectures of functional integument, particularly functional biomimetic dermal structures inclusive of cutaneous appendages. Although the tissue-engineered skin with complete biological activity and physiological functions is still cannot be manufactured, it is believed that with the advances in matrix materials, molding process, and biotechnology, a new generation of physiologically active skin will be born in the future. In pursuit of furnishing readers and researchers involved in relevant research to have a systematic and comprehensive understanding of 3D printed tissue-engineered skin, this paper furnishes an exegesis on the prevailing research landscape, formidable obstacles, and forthcoming trajectories within the sphere of tissue-engineered skin, including: (1) the prevalent biomaterials (collagen, chitosan, agarose, alginate, etc.) routinely employed in tissue-engineered skin, and a discerning analysis and comparison of their respective merits, demerits, and inherent characteristics; (2) the underlying principles and distinguishing attributes of various current printing methodologies utilized in tissue-engineered skin fabrication; (3) the present research status and progression in the realm of tissue-engineered biomimetic skin; (4) meticulous scrutiny and summation of the extant research underpinning tissue-engineered skin inform the identification of prevailing challenges and issues.
Collapse
Affiliation(s)
- Qinghua Wei
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China; Innovation Center NPU Chongqing, Northwestern Polytechnical University, Chongqing 400000, China.
| | - Yalong An
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Xudong Zhao
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Mingyang Li
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Juan Zhang
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
33
|
Westensee IN, Paffen LJMM, Pendlmayr S, De Dios Andres P, Ramos Docampo MA, Städler B. Artificial Cells and HepG2 Cells in 3D-Bioprinted Arrangements. Adv Healthc Mater 2024; 13:e2303699. [PMID: 38277695 DOI: 10.1002/adhm.202303699] [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: 11/27/2023] [Revised: 01/15/2024] [Indexed: 01/28/2024]
Abstract
Artificial cells are engineered units with cell-like functions for different purposes including acting as supportive elements for mammalian cells. Artificial cells with minimal liver-like function are made of alginate and equipped with metalloporphyrins that mimic the enzyme activity of a member of the cytochrome P450 family namely CYP1A2. The artificial cells are employed to enhance the dealkylation activity within 3D bioprinted structures composed of HepG2 cells and these artificial cells. This enhancement is monitored through the conversion of resorufin ethyl ether to resorufin. HepG2 cell aggregates are 3D bioprinted using an alginate/gelatin methacryloyl ink, resulting in the successful proliferation of the HepG2 cells. The composite ink made of an alginate/gelatin liquid phase with an increasing amount of artificial cells is characterized. The CYP1A2-like activity of artificial cells is preserved over at least 35 days, where 6 nM resorufin is produced in 8 h. Composite inks made of artificial cells and HepG2 cell aggregates in a liquid phase are used for 3D bioprinting. The HepG2 cells proliferate over 35 days, and the structure has boosted CYP1A2 activity. The integration of artificial cells and their living counterparts into larger 3D semi-synthetic tissues is a step towards exploring bottom-up synthetic biology in tissue engineering.
Collapse
Affiliation(s)
- Isabella N Westensee
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| | - Lars J M M Paffen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| | - Stefan Pendlmayr
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Paula De Dios Andres
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| | - Miguel A Ramos Docampo
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| | - Brigitte Städler
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| |
Collapse
|
34
|
Buckley C, Wang H, O'Dell R, Del Rosario M, Parimala Chelvi Ratnamani M, Rome M, Wang H. Creation of Porous, Perfusable Microtubular Networks for Improved Cell Viability in Volumetric Hydrogels. ACS APPLIED MATERIALS & INTERFACES 2024; 16:18522-18533. [PMID: 38564436 DOI: 10.1021/acsami.4c00716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The creation of large, volumetric tissue-engineered constructs has long been hindered due to the lack of effective vascularization strategies. Recently, 3D printing has emerged as a viable approach to creating vascular structures; however, its application is limited. Here, we present a simple and controllable technique to produce porous, free-standing, perfusable tubular networks from sacrificial templates of polyelectrolyte complex and coatings of salt-containing citrate-based elastomer poly(1,8-octanediol-co-citrate) (POC). As demonstrated, fully perfusable and interconnected POC tubular networks with channel diameters ranging from 100 to 400 μm were created. Incorporating NaCl particulates into the POC coating enabled the formation of micropores (∼19 μm in diameter) in the tubular wall upon particulate leaching to increase the cross-wall fluid transport. Casting and cross-linking gelatin methacrylate (GelMA) suspended with human osteoblasts over the free-standing porous POC tubular networks led to the fabrication of 3D cell-encapsulated constructs. Compared to the constructs without POC tubular networks, those with either solid or porous wall tubular networks exhibited a significant increase in cell viability and proliferation along with healthy cell morphology, particularly those with porous networks. Taken together, the sacrificial template-assisted approach is effective to fabricate tubular networks with controllable channel diameter and patency, which can be easily incorporated into cell-encapsulated hydrogels or used as tissue-engineering scaffolds to improve cell viability.
Collapse
Affiliation(s)
- Christian Buckley
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
- Semcer Center for Healthcare Innovation, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Haoyu Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
- Semcer Center for Healthcare Innovation, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Robert O'Dell
- Department of Chemical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Matthew Del Rosario
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Matangi Parimala Chelvi Ratnamani
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
- Semcer Center for Healthcare Innovation, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Mark Rome
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
- Semcer Center for Healthcare Innovation, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| |
Collapse
|
35
|
Jain P, Kathuria H, Ramakrishna S, Parab S, Pandey MM, Dubey N. In Situ Bioprinting: Process, Bioinks, and Applications. ACS APPLIED BIO MATERIALS 2024. [PMID: 38598256 DOI: 10.1021/acsabm.3c01303] [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: 04/11/2024]
Abstract
Traditional tissue engineering methods face challenges, such as fabrication, implantation of irregularly shaped scaffolds, and limited accessibility for immediate healthcare providers. In situ bioprinting, an alternate strategy, involves direct deposition of biomaterials, cells, and bioactive factors at the site, facilitating on-site fabrication of intricate tissue, which can offer a patient-specific personalized approach and align with the principles of precision medicine. It can be applied using a handled device and robotic arms to various tissues, including skin, bone, cartilage, muscle, and composite tissues. Bioinks, the critical components of bioprinting that support cell viability and tissue development, play a crucial role in the success of in situ bioprinting. This review discusses in situ bioprinting techniques, the materials used for bioinks, and their critical properties for successful applications. Finally, we discuss the challenges and future trends in accelerating in situ printing to translate this technology in a clinical settings for personalized regenerative medicine.
Collapse
Affiliation(s)
- Pooja Jain
- Faculty of Dentistry, National University of Singapore, Singapore 119805, Singapore
| | - Himanshu Kathuria
- Nusmetics Pte Ltd, E-Centre@Redhill, 3791 Jalan Bukit Merah, Singapore 159471, Singapore
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, Center for Nanotechnology and Sustainability, National University of Singapore, Singapore 117581, Singapore
| | - Shraddha Parab
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan India, 333031
| | - Murali M Pandey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan India, 333031
| | - Nileshkumar Dubey
- Faculty of Dentistry, National University of Singapore, Singapore 119805, Singapore
- ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore 119805, Singapore
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
Wang X, Zhang Y, Song A, Wang H, Wu Y, Chang W, Tian B, Xu J, Dai H, Ma Q, Wang C, Zhou X. A Printable Hydrogel Loaded with Medicinal Plant Extract for Promoting Wound Healing. Adv Healthc Mater 2024; 13:e2303017. [PMID: 38273733 DOI: 10.1002/adhm.202303017] [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: 09/09/2023] [Revised: 01/16/2024] [Indexed: 01/27/2024]
Abstract
How to promote wound healing is still a major challenge in the healthcare while macrophages are a critical component of the healing process. Compared to various bioactive drugs, many plants have been reported to facilitate the wound healing process by regulating the immune response of wounds. In this work, a Three-dimensional (3D) printed hydrogel scaffold loaded with natural Centella asiatica extract (CA extract) is developed for wound healing. This CA@3D scaffold uses gelatin (Gel) and sodium alginate (SA) with CA extract as bio-ink for 3D printing. The CA extract contains a variety of bioactive compounds that make the various active ingredients in Centella asiatica work in concert. The printed CA@3D scaffold can fit the shape of wound, orchestrate the macrophages and immune responses within the wound, and promote wound healing compared to commercial wound dressings. The underlying mechanism of promoting wound healing is also illuminated by applying multi-omic analyses. Moreover, the CA extract loaded 3D scaffold also showed great ability to promote wound healing in diabetic chronic wounds. Due to its ease of preparation, low-cost, biosafety, and therapeutic outcomes, this work proposes an effective strategy for promoting chronic wound healing.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yue Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Anning Song
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Heng Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yi Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Wenju Chang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, 233004, China
| | - Bo Tian
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Qingle Ma
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
39
|
Wu J, Yun Z, Song W, Yu T, Xue W, Liu Q, Sun X. Highly oriented hydrogels for tissue regeneration: design strategies, cellular mechanisms, and biomedical applications. Theranostics 2024; 14:1982-2035. [PMID: 38505623 PMCID: PMC10945336 DOI: 10.7150/thno.89493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/19/2024] [Indexed: 03/21/2024] Open
Abstract
Many human tissues exhibit a highly oriented architecture that confers them with distinct mechanical properties, enabling adaptation to diverse and challenging environments. Hydrogels, with their water-rich "soft and wet" structure, have emerged as promising biomimetic materials in tissue engineering for repairing and replacing damaged tissues and organs. Highly oriented hydrogels can especially emulate the structural orientation found in human tissue, exhibiting unique physiological functions and properties absent in traditional homogeneous isotropic hydrogels. The design and preparation of highly oriented hydrogels involve strategies like including hydrogels with highly oriented nanofillers, polymer-chain networks, void channels, and microfabricated structures. Understanding the specific mechanism of action of how these highly oriented hydrogels affect cell behavior and their biological applications for repairing highly oriented tissues such as the cornea, skin, skeletal muscle, tendon, ligament, cartilage, bone, blood vessels, heart, etc., requires further exploration and generalization. Therefore, this review aims to fill that gap by focusing on the design strategy of highly oriented hydrogels and their application in the field of tissue engineering. Furthermore, we provide a detailed discussion on the application of highly oriented hydrogels in various tissues and organs and the mechanisms through which highly oriented structures influence cell behavior.
Collapse
Affiliation(s)
- Jiuping Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhihe Yun
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wenlong Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130023, China
| | - Tao Yu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wu Xue
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Qinyi Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Xinzhi Sun
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
40
|
Chowdhury SR, Mondal G, Ratnayake P, Basu B. Three-Dimensional Extrusion Printed Urinary Specific Grafts: Mechanistic Insights into Buildability and Biophysical Properties. ACS Biomater Sci Eng 2024; 10:1040-1061. [PMID: 38294204 DOI: 10.1021/acsbiomaterials.3c01422] [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: 02/01/2024]
Abstract
The compositional formulations and the optimization of process parameters to fabricate hydrogel scaffolds with urological tissue-mimicking biophysical properties are not yet extensively explored, including a comprehensive assessment of a spectrum of properties, such as mechanical strength, viscoelasticity, antimicrobial property, and cytocompatibility. While addressing this aspect, the present work provides mechanistic insights into process science, to produce shape-fidelity compliant alginate-based biomaterial ink blended with gelatin and synthetic nanocellulose. The composition-dependent pseudoplasticity, viscoelasticity, thixotropy, and gel stability over a longer duration in physiological context have been rationalized in terms of intermolecular hydrogen bonding interactions among the biomaterial ink constituents. By varying the hybrid hydrogel ink composition within a narrow compositional window, the resulting hydrogel closely mimics the natural urological tissue-like properties, including tensile stretchability, compressive strength, and biophysical properties. Based on the printability assessment using a critical analysis of gel strength, we have established the buildability of the acellular hydrogel ink and have been successful in fabricating shape-fidelity compliant urological patches or hollow cylindrical grafts using 3D extrusion printing. Importantly, the new hydrogel formulations with good hydrophilicity, support fibroblast cell proliferation and inhibit the growth of Gram-negative E. coli bacteria. These attributes were rationalized in terms of nanocellulose-induced physicochemical changes on the scaffold surface. Taken together, the present study uncovers the process-science-based understanding of the 3D extrudability of the newly formulated alginate-gelatin-nanocellulose-based hydrogels with urological tissue-specific biophysical, cytocompatibility, and antibacterial properties.
Collapse
Affiliation(s)
- Sulob Roy Chowdhury
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Garga Mondal
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Praneeth Ratnayake
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
41
|
Kalogeropoulou M, Díaz-Payno PJ, Mirzaali MJ, van Osch GJVM, Fratila-Apachitei LE, Zadpoor AA. 4D printed shape-shifting biomaterials for tissue engineering and regenerative medicine applications. Biofabrication 2024; 16:022002. [PMID: 38224616 DOI: 10.1088/1758-5090/ad1e6f] [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: 08/30/2023] [Accepted: 01/15/2024] [Indexed: 01/17/2024]
Abstract
The existing 3D printing methods exhibit certain fabrication-dependent limitations for printing curved constructs that are relevant for many tissues. Four-dimensional (4D) printing is an emerging technology that is expected to revolutionize the field of tissue engineering and regenerative medicine (TERM). 4D printing is based on 3D printing, featuring the introduction of time as the fourth dimension, in which there is a transition from a 3D printed scaffold to a new, distinct, and stable state, upon the application of one or more stimuli. Here, we present an overview of the current developments of the 4D printing technology for TERM, with a focus on approaches to achieve temporal changes of the shape of the printed constructs that would enable biofabrication of highly complex structures. To this aim, the printing methods, types of stimuli, shape-shifting mechanisms, and cell-incorporation strategies are critically reviewed. Furthermore, the challenges of this very recent biofabrication technology as well as the future research directions are discussed. Our findings show that the most common printing methods so far are stereolithography (SLA) and extrusion bioprinting, followed by fused deposition modelling, while the shape-shifting mechanisms used for TERM applications are shape-memory and differential swelling for 4D printing and 4D bioprinting, respectively. For shape-memory mechanism, there is a high prevalence of synthetic materials, such as polylactic acid (PLA), poly(glycerol dodecanoate) acrylate (PGDA), or polyurethanes. On the other hand, different acrylate combinations of alginate, hyaluronan, or gelatin have been used for differential swelling-based 4D transformations. TERM applications include bone, vascular, and cardiac tissues as the main target of the 4D (bio)printing technology. The field has great potential for further development by considering the combination of multiple stimuli, the use of a wider range of 4D techniques, and the implementation of computational-assisted strategies.
Collapse
Affiliation(s)
- Maria Kalogeropoulou
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Delft, CD 2628, The Netherlands
| | - Pedro J Díaz-Payno
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Delft, CD 2628, The Netherlands
- Department of Orthopedics and Sports Medicine, Erasmus MC University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Mohammad J Mirzaali
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Delft, CD 2628, The Netherlands
| | - Gerjo J V M van Osch
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Delft, CD 2628, The Netherlands
- Department of Orthopedics and Sports Medicine, Erasmus MC University Medical Center, 3015 CN Rotterdam, The Netherlands
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Lidy E Fratila-Apachitei
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Delft, CD 2628, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Delft, CD 2628, The Netherlands
- Department of Orthopedics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
42
|
Sheng L, Song X, Wang M, Zheng S. Thermally reversible hydrogels printing of customizable bio-channels with curvature. Int J Biol Macromol 2024; 257:128595. [PMID: 38056748 DOI: 10.1016/j.ijbiomac.2023.128595] [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: 10/18/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/08/2023]
Abstract
Replicating intricate bio-channels, akin to expansive vascular networks, offers numerous advantages including self-repair, replacing damaged bio-channels, testing drugs, and biomedical devices. But, crafting multi-sized, editable bio-channels with specific curvatures, particularly using natural polymer-based bio-inks, poses a significant challenge. To address this, this study introduces a temperature-driven indirect printing method, exemplified by the diploic vein. Here, K-carrageenan (kca)-silk fiber (SF)-hyaluronic acid (HA)/hFOB 1.19 (SV40 transfection of human osteoblasts) and kca-collagen-HA/HUVECs (human umbilical vein endothelial cells) are employed to fabricate vascular-like walls and lumens, utilizing their thermoreversible properties to create multi-stage bifurcated lumens. Precise spatial curvature was generated by heating the vascular network wrapped in poly(N-isopropyl acrylamide) (PNIPAAm)-poly(ethylene glycol) diacrylate (PEGDA). Since temperature is specific to the thermal material carrying the cells, the rheological properties of bioinks, modeling temperature parameters, and their impact on printing size was explored. Additionally, mechanical properties and curvature response were characterized to determine the necessary process parameters for achieving the desired size. Ultimately, in vitro bioprinting experiments involving HUVECs and hFOB 1.19 demonstrate cell viability, adhesion, proliferation, and migration within the intraluminal hydrogel scaffold. This approach allows for customizing bio-channel content and controlling curvature programming, providing new prospects for in vitro biochannel production, with potential benefits for pathology research.
Collapse
Affiliation(s)
- Lin Sheng
- Tianjin Key Laboratory of Equipment Design and Manufacturing Technology, School of Mechanical Engineering, Tianjin University, Tianjin 300354, China
| | - Xiaofei Song
- Tianjin Key Laboratory of Equipment Design and Manufacturing Technology, School of Mechanical Engineering, Tianjin University, Tianjin 300354, China
| | - Miaomiao Wang
- Tianjin Key Laboratory of Equipment Design and Manufacturing Technology, School of Mechanical Engineering, Tianjin University, Tianjin 300354, China
| | - Shuxian Zheng
- Tianjin Key Laboratory of Equipment Design and Manufacturing Technology, School of Mechanical Engineering, Tianjin University, Tianjin 300354, China.
| |
Collapse
|
43
|
Lv J, Du X, Wang M, Su J, Wei Y, Xu C. Construction of tumor organoids and their application to cancer research and therapy. Theranostics 2024; 14:1101-1125. [PMID: 38250041 PMCID: PMC10797287 DOI: 10.7150/thno.91362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Cancer remains a severe public health burden worldwide. One of the challenges hampering effective cancer therapy is that the existing cancer models hardly recapitulate the tumor microenvironment of human patients. Over the past decade, tumor organoids have emerged as an in vitro 3D tumor model to mimic the pathophysiological characteristics of parental tumors. Various techniques have been developed to construct tumor organoids, such as matrix-based methods, hanging drop, spinner or rotating flask, nonadhesive surface, organ-on-a-chip, 3D bioprinting, and genetic engineering. This review elaborated on cell components and fabrication methods for establishing tumor organoid models. Furthermore, we discussed the application of tumor organoids to cancer modeling, basic cancer research, and anticancer therapy. Finally, we discussed current limitations and future directions in employing tumor organoids for more extensive applications.
Collapse
Affiliation(s)
- Jiajing Lv
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Institute of Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Xuan Du
- Biopharma Industry Promotion Center Shanghai, Shanghai 201203, China
| | - Miaomiao Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Institute of Medicine, Shanghai University, Shanghai 200444, China
- Department of Rehabilitation Medicine, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Can Xu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
44
|
Thangadurai M, Srinivasan SS, Sekar MP, Sethuraman S, Sundaramurthi D. Emerging perspectives on 3D printed bioreactors for clinical translation of engineered and bioprinted tissue constructs. J Mater Chem B 2024; 12:350-381. [PMID: 38084021 DOI: 10.1039/d3tb01847d] [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: 01/05/2024]
Abstract
3D printed/bioprinted tissue constructs are utilized for the regeneration of damaged tissues and as in vitro models. Most of the fabricated 3D constructs fail to undergo functional maturation in conventional in vitro settings. There is a challenge to provide a suitable niche for the fabricated tissue constructs to undergo functional maturation. Bioreactors have emerged as a promising tool to enhance tissue maturation of the engineered constructs by providing physical/biological cues along with a controlled nutrient supply under dynamic in vitro conditions. Bioreactors provide an ambient microenvironment most appropriate for the development of functionally matured tissue constructs by promoting cell proliferation, differentiation, and maturation for transplantation and drug screening applications. Due to the huge cost and limited availability of commercial bioreactors, there is a need to develop strategies to make customized bioreactors. Additive manufacturing (AM) may be a viable tool to fabricate custom designed bioreactors with better efficiency and at low cost. In this review, we have extensively discussed the importance of bioreactors in functionalizing tissue engineered/3D bioprinted scaffolds for bone, cartilage, skeletal muscle, nerve, and vascular tissue. In addition, the importance and fabrication of customized 3D printed bioreactors for the maturation of tissue engineered constructs are discussed in detail. Finally, the current challenges and future perspectives in translating commercial and custom 3D printed bioreactors for clinical applications are outlined.
Collapse
Affiliation(s)
- Madhumithra Thangadurai
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| | - Sai Sadhananth Srinivasan
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| | - Muthu Parkkavi Sekar
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| |
Collapse
|
45
|
Sun H, Sun L, Ke X, Liu L, Li C, Jin B, Wang P, Jiang Z, Zhao H, Yang Z, Sun Y, Liu J, Wang Y, Sun M, Pang M, Wang Y, Wu B, Zhao H, Sang X, Xing B, Yang H, Huang P, Mao Y. Prediction of Clinical Precision Chemotherapy by Patient-Derived 3D Bioprinting Models of Colorectal Cancer and Its Liver Metastases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304460. [PMID: 37973557 PMCID: PMC10787059 DOI: 10.1002/advs.202304460] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/29/2023] [Indexed: 11/19/2023]
Abstract
Methods accurately predicting the responses of colorectal cancer (CRC) and colorectal cancer liver metastasis (CRLM) to personalized chemotherapy remain limited due to tumor heterogeneity. This study introduces an innovative patient-derived CRC and CRLM tumor model for preclinical investigation, utilizing 3d-bioprinting (3DP) technology. Efficient construction of homogeneous in vitro 3D models of CRC/CRLM is achieved through the application of patient-derived primary tumor cells and 3D bioprinting with bioink. Genomic and histological analyses affirm that the CRC/CRLM 3DP tumor models effectively retain parental tumor biomarkers and mutation profiles. In vitro tests evaluating chemotherapeutic drug sensitivities reveal substantial tumor heterogeneity in chemotherapy responses within the 3DP CRC/CRLM models. Furthermore, a robust correlation is evident between the drug response in the CRLM 3DP model and the clinical outcomes of neoadjuvant chemotherapy. These findings imply a significant potential for the application of patient-derived 3DP cancer models in precision chemotherapy prediction and preclinical research for CRC/CRLM.
Collapse
Affiliation(s)
- Hang Sun
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Lejia Sun
- Department of General SurgeryThe First Affiliated HospitalNanjing Medical UniversityNanjingJiangsu210029China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Xindi Ke
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Lijuan Liu
- Department of Hepatopancreatobiliary Surgery IKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijing100142China
| | - Changcan Li
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Bao Jin
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Peipei Wang
- Department of General SurgeryThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Zhuoran Jiang
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Hong Zhao
- Department of Hepatobiliary SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Zhiying Yang
- First Department of Hepatopancreatobiliary SurgeryChina‐Japan Friendship HospitalBeijing100029China
| | - Yongliang Sun
- First Department of Hepatopancreatobiliary SurgeryChina‐Japan Friendship HospitalBeijing100029China
| | - Jianmei Liu
- Department of Hepatobiliary SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Yan Wang
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Minghao Sun
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Mingchang Pang
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Yinhan Wang
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Bin Wu
- Department of General SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Haitao Zhao
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Xinting Sang
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Baocai Xing
- Department of Hepatopancreatobiliary Surgery IKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijing100142China
| | - Huayu Yang
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| | - Pengyu Huang
- State Key Laboratory of Advanced Medical Materials and DevicesEngineering Research Center of Pulmonary and Critical Care Medicine Technology and Device (Ministry of Education)Institute of Biomedical EngineeringChinese Academy of Medical Science & Peking Union Medical CollegeTianjin300192China
- Tianjin Institutes of Health ScienceTianjin301600China
| | - Yilei Mao
- Department of Liver SurgeryPeking Union Medical College (PUMC) HospitalPeking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS)Beijing100730China
| |
Collapse
|
46
|
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
|
47
|
Heidari F, Saadatmand M, Simorgh S. Directly coaxial bioprinting of 3D vascularized tissue using novel bioink based on decellularized human amniotic membrane. Int J Biol Macromol 2023; 253:127041. [PMID: 37742904 DOI: 10.1016/j.ijbiomac.2023.127041] [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: 07/20/2023] [Revised: 09/05/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Despite several progressions in the biofabrication of large-scale engineered tissues, direct biopri nting of perfusable three-dimensional (3D) vasculature remained unaddressed. Developing a feasible method to generate cell-laden thick tissue with an effective vasculature network to deliver oxygen and nutrient is crucial for preventing the formation of necrotic spots and tissue death. In this study, we developed a novel technique to directly bioprint 3D cell-laden prevascularized construct. We developed a novel bioink by mixing decellularized human amniotic membrane (dHAM) and alginate (Alg) in various ratios. The bioink with encapsulated human vein endothelial cells (HUVECs) and a crosslinker, CaCl2, were extruded via sheath and core nozzle respectively to directly bioprint a perfusable 3D vasculature construct. The various concentration of bioink was assessed from several aspects like biocompatibility, porosity, swelling, degradation, and mechanical characteristics, and accordingly, optimized concentration was selected (Alg 4 %w/v - dHAM 0.6 %w/v). Then, the crosslinked bioink without microchannel and the 3D bioprinted construct with various microchannel distances (0, 1.5 mm, 3 mm) were compared. The 3D bioprinted construct with a 1.5 mm microchannels distance demonstrated superiority owing to its 492 ± 18.8 % cell viability within 14 days, excellent tubulogenesis, remarkable expression of VEGFR-2 which play a crucial role in endothelial cell proliferation, migration, and more importantly angiogenesis, and neovascularization. This perfusable bioprinted construct also possess appropriate mechanical stability (32.35 ± 5 kPa Young's modulus) for soft tissue. Taking these advantages into the account, our new bioprinting method possesses a prominent potential for the fabrication of large-scale prevascularized tissue to serve for regenerative medicine applications like implantation, drug-screening platform, and the study of mutation disease.
Collapse
Affiliation(s)
- Faranak Heidari
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | - Maryam Saadatmand
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | - Sara Simorgh
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Verma S, Khanna V, Kumar S, Kumar S. The Art of Building Living Tissues: Exploring the Frontiers of Biofabrication with 3D Bioprinting. ACS OMEGA 2023; 8:47322-47339. [PMID: 38144142 PMCID: PMC10734012 DOI: 10.1021/acsomega.3c02600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 09/11/2023] [Indexed: 12/26/2023]
Abstract
The scope of three-dimensional printing is expanding rapidly, with innovative approaches resulting in the evolution of state-of-the-art 3D bioprinting (3DbioP) techniques for solving issues in bioengineering and biopharmaceutical research. The methods and tools in 3DbioP emphasize the extrusion process, bioink formulation, and stability of the bioprinted scaffold. Thus, 3DbioP technology augments 3DP in the biological world by providing technical support to regenerative therapy, drug delivery, bioengineering of prosthetics, and drug kinetics research. Besides the above, drug delivery and dosage control have been achieved using 3D bioprinted microcarriers and capsules. Developing a stable, biocompatible, and versatile bioink is a primary requisite in biofabrication. The 3DbioP research is breaking the technical barriers at a breakneck speed. Numerous techniques and biomaterial advancements have helped to overcome current 3DbioP issues related to printability, stability, and bioink formulation. Therefore, this Review aims to provide an insight into the technical challenges of bioprinting, novel biomaterials for bioink formulation, and recently developed 3D bioprinting methods driving future applications in biofabrication research.
Collapse
Affiliation(s)
- Saurabh Verma
- Department
of Health Research-Multi-Disciplinary Research Unit, King George’s Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Vikram Khanna
- Department
of Oral Medicine and Radiology, King George’s
Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Smita Kumar
- Department
of Health Research-Multi-Disciplinary Research Unit, King George’s Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Sumit Kumar
- Department
of Health Research-Multi-Disciplinary Research Unit, King George’s Medical University, Lucknow, Uttar Pradesh 226003, India
| |
Collapse
|
49
|
Shin S, Brunel LG, Cai B, Kilian D, Roth JG, Seymour AJ, Heilshorn SC. Gelation of Uniform Interfacial Diffusant in Embedded 3D Printing. ADVANCED FUNCTIONAL MATERIALS 2023; 33:2307435. [PMID: 38646474 PMCID: PMC11031202 DOI: 10.1002/adfm.202307435] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Indexed: 04/23/2024]
Abstract
While the human body has many different examples of perfusable structures with complex geometries, biofabrication methods to replicate this complexity are still lacking. Specifically, the fabrication of self-supporting, branched networks with multiple channel diameters is particularly challenging. Here, we present the Gelation of Uniform Interfacial Diffusant in Embedded 3D Printing (GUIDE-3DP) approach for constructing perfusable networks of interconnected channels with precise control over branching geometries and vessel sizes. To achieve user-specified channel dimensions, this technique leverages the predictable diffusion of crosslinking reaction-initiators released from sacrificial inks printed within a hydrogel precursor. We demonstrate the versatility of GUIDE-3DP to be adapted for use with diverse physicochemical crosslinking mechanisms by designing seven printable material systems. Importantly, GUIDE-3DP allows for the independent tunability of both the inner and outer diameters of the printed channels and the ability to fabricate seamless junctions at branch points. This 3D bioprinting platform is uniquely suited for fabricating lumenized structures with complex shapes characteristic of multiple hollow vessels throughout the body. As an exemplary application, we demonstrate the fabrication of vasculature-like networks lined with endothelial cells. GUIDE-3DP represents an important advance toward the fabrication of self-supporting, physiologically relevant networks with intricate and perfusable geometries.
Collapse
Affiliation(s)
- Sungchul Shin
- Department of Materials Science and Engineering, Stanford University, 466 Lomita Mall, Stanford, CA 94305, USA
- Department of Agriculture, Forestry, and Bioresources, Seoul National University, 08826 Gwanak-ro 1, Gwanak-gu, Seoul, Republic of Korea
| | - Lucia G Brunel
- Department of Chemical Engineering, Stanford University, 466 Lomita Mall, Stanford, CA 94305, USA
| | - Betty Cai
- Department of Materials Science and Engineering, Stanford University, 466 Lomita Mall, Stanford, CA 94305, USA
| | - David Kilian
- Department of Materials Science and Engineering, Stanford University, 466 Lomita Mall, Stanford, CA 94305, USA
| | - Julien G Roth
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, 466 Lomita Mall, Stanford, CA 94305, USA
| | - Alexis J Seymour
- Department of Bioengineering, Stanford University, 466 Lomita Mall, Stanford, CA 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, 466 Lomita Mall, Stanford, CA 94305, USA
| |
Collapse
|
50
|
Wei SY, Chen PY, Hsieh CC, Chen YS, Chen TH, Yu YS, Tsai MC, Xie RH, Chen GY, Yin GC, Melero-Martin JM, Chen YC. Engineering large and geometrically controlled vascularized nerve tissue in collagen hydrogels to restore large-sized volumetric muscle loss. Biomaterials 2023; 303:122402. [PMID: 37988898 DOI: 10.1016/j.biomaterials.2023.122402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023]
Abstract
Developing scalable vascularized and innervated tissue is a critical challenge for the successful clinical application of tissue-engineered constructs. Collagen hydrogels are extensively utilized in cell-mediated vascular network formation because of their naturally excellent biological properties. However, the substantial increase in hydrogel contraction induced by populated cells limits their long-term use. Previous studies attempted to mitigate this issue by concentrating collagen pre-polymer solutions or synthesizing covalently crosslinked collagen hydrogels. However, these methods only partially reduce hydrogel contraction while hindering blood vessel formation within the hydrogels. To address this challenge, we introduced additional support in the form of a supportive spacer to counteract the contraction forces of populated cells and prevent hydrogel contraction. This approach was found to promote cell spreading, resist hydrogel contraction, control hydrogel/tissue geometry, and even facilitate the engineering of functional blood vessels and host nerve growth in just one week. Subsequently, implanting these engineered tissues into muscle defect sites resulted in timely anastomosis with the host vasculature, leading to enhanced myogenesis, increased muscle innervation, and the restoration of injured muscle functionality. Overall, this innovative strategy expands the applicability of collagen hydrogels in fabricating large vascularized nerve tissue constructs for repairing volumetric muscle loss (∼63 %) and restoring muscle function.
Collapse
Affiliation(s)
- Shih-Yen Wei
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Po-Yu Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Chia-Chang Hsieh
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Yu-Shan Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Tzu-Hsuan Chen
- Department of Materials Science and Engineering, Carnegie Mellon University, PA, USA
| | - Yu-Shan Yu
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Min-Chun Tsai
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Ren-Hao Xie
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Guan-Yu Chen
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Gung-Chian Yin
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Ying-Chieh Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan.
| |
Collapse
|