1
|
Yilmaz G. Foundational Engineering of Artificial Blood Vessels' Biomechanics: The Impact of Wavy Geometric Designs. Biomimetics (Basel) 2024; 9:546. [PMID: 39329568 PMCID: PMC11430736 DOI: 10.3390/biomimetics9090546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/21/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
The design of wavy structures and their mechanical implications on artificial blood vessels (ABVs) have been insufficiently studied in the existing literature. This research aims to explore the influence of various wavy geometric designs on the mechanical properties of ABVs and to establish a foundational framework for advancing and applying these designs. Computer-aided design (CAD) and finite element method (FEM) simulations, in conjunction with physical sample testing, were utilized. A geometric model incorporating concave and convex curves was developed and analyzed with a symbolic mathematical tool. Subsequently, a total of ten CAD models were subjected to increasing internal pressures using a FEM simulation to evaluate the expansion of internal areas. Additionally, physical experiments were conducted further to investigate the expansion of ABV samples under pressure. The results demonstrated that increased wave numbers significantly enhance the flexibility of ABVs. Samples with 22 waves exhibited a 45% larger area under 24 kPa pressure than those with simple circles. However, the increased number of waves also led to undesirable high-pressure gradients at elevated pressures. Furthermore, a strong correlation was observed between the experimental outcomes and the simulation results, with a notably low error margin, ranging from 19.88% to 3.84%. Incorporating wavy designs into ABVs can effectively increase both vessel flexibility and the internal area under pressure. Finally, it was found that expansion depending on the wave number can be efficiently modeled with a simple linear equation, which could be utilized in future designs.
Collapse
Affiliation(s)
- Galip Yilmaz
- Electronics and Automation Department, Bayburt University, Bayburt 69000, Turkey
| |
Collapse
|
2
|
Nain A, Chakraborty S, Barman SR, Gavit P, Indrakumar S, Agrawal A, Lin ZH, Chatterjee K. Progress in the development of piezoelectric biomaterials for tissue remodeling. Biomaterials 2024; 307:122528. [PMID: 38522326 DOI: 10.1016/j.biomaterials.2024.122528] [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: 11/02/2023] [Revised: 02/15/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
Piezoelectric biomaterials have demonstrated significant potential in the past few decades to heal damaged tissue and restore cellular functionalities. Herein, we discuss the role of bioelectricity in tissue remodeling and explore ways to mimic such tissue-like properties in synthetic biomaterials. In the past decade, biomedical engineers have adopted emerging functional biomaterials-based tissue engineering approaches using innovative bioelectronic stimulation protocols based on dynamic stimuli to direct cellular activation, proliferation, and differentiation on engineered biomaterial constructs. The primary focus of this review is to discuss the concepts of piezoelectric energy harvesting, piezoelectric materials, and their application in soft (skin and neural) and hard (dental and bone) tissue regeneration. While discussing the prospective applications as an engineered tissue, an important distinction has been made between piezoceramics, piezopolymers, and their composites. The superiority of piezopolymers over piezoceramics to circumvent issues such as stiffness mismatch, biocompatibility, and biodegradability are highlighted. We aim to provide a comprehensive review of the field and identify opportunities for the future to develop clinically relevant and state-of-the-art biomaterials for personalized and remote health care.
Collapse
Affiliation(s)
- Amit Nain
- Department of Material Engineering, Indian Institute of Science, Bangalore, 560012, Karnataka, India.
| | - Srishti Chakraborty
- Department of Material Engineering, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Snigdha Roy Barman
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Pratik Gavit
- Department of Material Engineering, Indian Institute of Science, Bangalore, 560012, Karnataka, India; School of Bio Science and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Sushma Indrakumar
- Department of Material Engineering, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Akhilesh Agrawal
- Department of Material Engineering, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Zong-Hong Lin
- Department of Biomedical Engineering, National Taiwan University, Taipe, 10617, Taiwan.
| | - Kaushik Chatterjee
- Department of Material Engineering, Indian Institute of Science, Bangalore, 560012, Karnataka, India; Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, Karnataka, India.
| |
Collapse
|
3
|
Zheng Z, Tang W, Li Y, Ai Y, Tu Z, Yang J, Fan C. Advancing cardiac regeneration through 3D bioprinting: methods, applications, and future directions. Heart Fail Rev 2024; 29:599-613. [PMID: 37943420 DOI: 10.1007/s10741-023-10367-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2023] [Indexed: 11/10/2023]
Abstract
Cardiovascular diseases (CVDs) represent a paramount global mortality concern, and their prevalence is on a relentless ascent. Despite the effectiveness of contemporary medical interventions in mitigating CVD-related fatality rates and complications, their efficacy remains curtailed by an array of limitations. These include the suboptimal efficiency of direct cell injection and an inherent disequilibrium between the demand and availability of heart transplantations. Consequently, the imperative to formulate innovative strategies for cardiac regeneration therapy becomes unmistakable. Within this context, 3D bioprinting technology emerges as a vanguard contender, occupying a pivotal niche in the realm of tissue engineering and regenerative medicine. This state-of-the-art methodology holds the potential to fabricate intricate heart tissues endowed with multifaceted structures and functionalities, thereby engendering substantial promise. By harnessing the prowess of 3D bioprinting, it becomes plausible to synthesize functional cardiac architectures seamlessly enmeshed with the host tissue, affording a viable avenue for the restitution of infarcted domains and, by extension, mitigating the onerous yoke of CVDs. In this review, we encapsulate the myriad applications of 3D bioprinting technology in the domain of heart tissue regeneration. Furthermore, we usher in the latest advancements in printing methodologies and bioinks, culminating in an exploration of the extant challenges and the vista of possibilities inherent to a diverse array of approaches.
Collapse
Affiliation(s)
- Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Weijie Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Yichen Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Yinze Ai
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Zhi Tu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Jinfu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China.
| |
Collapse
|
4
|
Laowpanitchakorn P, Zeng J, Piantino M, Uchida K, Katsuyama M, Matsusaki M. Biofabrication of engineered blood vessels for biomedical applications. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2330339. [PMID: 38633881 PMCID: PMC11022926 DOI: 10.1080/14686996.2024.2330339] [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: 12/12/2023] [Accepted: 03/10/2024] [Indexed: 04/19/2024]
Abstract
To successfully engineer large-sized tissues, establishing vascular structures is essential for providing oxygen, nutrients, growth factors and cells to prevent necrosis at the core of the tissue. The diameter scale of the biofabricated vasculatures should range from 100 to 1,000 µm to support the mm-size tissue while being controllably aligned and spaced within the diffusion limit of oxygen. In this review, insights regarding biofabrication considerations and techniques for engineered blood vessels will be presented. Initially, polymers of natural and synthetic origins can be selected, modified, and combined with each other to support maturation of vascular tissue while also being biocompatible. After they are shaped into scaffold structures by different fabrication techniques, surface properties such as physical topography, stiffness, and surface chemistry play a major role in the endothelialization process after transplantation. Furthermore, biological cues such as growth factors (GFs) and endothelial cells (ECs) can be incorporated into the fabricated structures. As variously reported, fabrication techniques, especially 3D printing by extrusion and 3D printing by photopolymerization, allow the construction of vessels at a high resolution with diameters in the desired range. Strategies to fabricate of stable tubular structures with defined channels will also be discussed. This paper provides an overview of the many advances in blood vessel engineering and combinations of different fabrication techniques up to the present time.
Collapse
Affiliation(s)
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Kentaro Uchida
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Misa Katsuyama
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
5
|
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
|
6
|
Choi J, Lee EJ, Jang WB, Kwon SM. Development of Biocompatible 3D-Printed Artificial Blood Vessels through Multidimensional Approaches. J Funct Biomater 2023; 14:497. [PMID: 37888162 PMCID: PMC10607080 DOI: 10.3390/jfb14100497] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
Within the human body, the intricate network of blood vessels plays a pivotal role in transporting nutrients and oxygen and maintaining homeostasis. Bioprinting is an innovative technology with the potential to revolutionize this field by constructing complex multicellular structures. This technique offers the advantage of depositing individual cells, growth factors, and biochemical signals, thereby facilitating the growth of functional blood vessels. Despite the challenges in fabricating vascularized constructs, bioprinting has emerged as an advance in organ engineering. The continuous evolution of bioprinting technology and biomaterial knowledge provides an avenue to overcome the hurdles associated with vascularized tissue fabrication. This article provides an overview of the biofabrication process used to create vascular and vascularized constructs. It delves into the various techniques used in vascular engineering, including extrusion-, droplet-, and laser-based bioprinting methods. Integrating these techniques offers the prospect of crafting artificial blood vessels with remarkable precision and functionality. Therefore, the potential impact of bioprinting in vascular engineering is significant. With technological advances, it holds promise in revolutionizing organ transplantation, tissue engineering, and regenerative medicine. By mimicking the natural complexity of blood vessels, bioprinting brings us one step closer to engineering organs with functional vasculature, ushering in a new era of medical advancement.
Collapse
Affiliation(s)
- Jaewoo Choi
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Eun Ji Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
7
|
Pandey A, Pragya, Kanoujia J, Parashar P. New Insights into the Applications of 3D-Printed Biomaterial in Wound Healing and Prosthesis. AAPS PharmSciTech 2023; 24:191. [PMID: 37726576 DOI: 10.1208/s12249-023-02643-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023] Open
Abstract
Recently three-dimensional bioprinting (3D-bioP) has emerged as a revolutionary technique for numerous biomedical applications. 3D-bioP has facilitated the printing of advanced and complex human organs resulting in satisfactory therapeutic practice. One of the important biomedical applications of 3D-bioP is in tissue engineering, wound healing, and prosthetics. 3D-bioP is basically aimed to restore the natural extracellular matrix of human's damage due to wounds. The relevant search was explored using various scientific database, viz., PubMed, Web of Science, Scopus, and ScienceDirect. The objective of this review is to emphasize interpretations from the pre-executed studies and to assess the worth of employing 3D-bioP in wound healing as well as prosthetics in terms of patient compliance, clinical outcomes, and economic viability. Furthermore, the benefits of applying 3D-bioP in wound healing over traditional methods have been covered along with the biocompatible biomaterials employed as bioinks has been discussion. Additionally, the review expands about the clinical trials in 3D-bioP field, showing promise of biomedical applicability of this technique with growing advancement in recent years.
Collapse
Affiliation(s)
- Aayushi Pandey
- Amity Institute of Pharmacy, Amity University Uttar Pradesh Lucknow Campus, Lucknow, U.P., 226028, India
| | - Pragya
- Amity Institute of Pharmacy, Amity University Uttar Pradesh Lucknow Campus, Lucknow, U.P., 226028, India
| | - Jovita Kanoujia
- Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, Madhya Pradesh, 474005, India
| | - Poonam Parashar
- Amity Institute of Pharmacy, Amity University Uttar Pradesh Lucknow Campus, Lucknow, U.P., 226028, India.
| |
Collapse
|
8
|
Yang Q, Li M, Yang X, Xiao Z, Tong X, Tuerdi A, Li S, Lei L. Flourishing tumor organoids: History, emerging technology, and application. Bioeng Transl Med 2023; 8:e10559. [PMID: 37693042 PMCID: PMC10487342 DOI: 10.1002/btm2.10559] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/16/2023] [Accepted: 05/25/2023] [Indexed: 09/12/2023] Open
Abstract
Malignant tumors are one of the leading causes of death which impose an increasingly heavy burden on all countries. Therefore, the establishment of research models that closely resemble original tumor characteristics is crucial to further understanding the mechanisms of malignant tumor development, developing safer and more effective drugs, and formulating personalized treatment plans. Recently, organoids have been widely used in tumor research owing to their advantages including preserving the structure, heterogeneity, and cellular functions of the original tumor, together with the ease of manipulation. This review describes the history and characteristics of tumor organoids and the synergistic combination of three-dimensional (3D) culture approaches for tumor organoids with emerging technologies, including tissue-engineered cell scaffolds, microfluidic devices, 3D bioprinting, rotating wall vessels, and clustered regularly interspaced short palindromic repeats-CRISPR-associated protein 9 (CRISPR-Cas9). Additionally, the progress in research and the applications in basic and clinical research of tumor organoid models are summarized. This includes studies of the mechanism of tumor development, drug development and screening, precision medicine, immunotherapy, and simulation of the tumor microenvironment. Finally, the existing shortcomings of tumor organoids and possible future directions are discussed.
Collapse
Affiliation(s)
- Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zian Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ayinuer Tuerdi
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| |
Collapse
|
9
|
Afewerki S, Stocco TD, Rosa da Silva AD, Aguiar Furtado AS, Fernandes de Sousa G, Ruiz-Esparza GU, Webster TJ, Marciano FR, Strømme M, Zhang YS, Lobo AO. In vitro high-content tissue models to address precision medicine challenges. Mol Aspects Med 2023; 91:101108. [PMID: 35987701 PMCID: PMC9384546 DOI: 10.1016/j.mam.2022.101108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/29/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2023]
Abstract
The field of precision medicine allows for tailor-made treatments specific to a patient and thereby improve the efficiency and accuracy of disease prevention, diagnosis, and treatment and at the same time would reduce the cost, redundant treatment, and side effects of current treatments. Here, the combination of organ-on-a-chip and bioprinting into engineering high-content in vitro tissue models is envisioned to address some precision medicine challenges. This strategy could be employed to tackle the current coronavirus disease 2019 (COVID-19), which has made a significant impact and paradigm shift in our society. Nevertheless, despite that vaccines against COVID-19 have been successfully developed and vaccination programs are already being deployed worldwide, it will likely require some time before it is available to everyone. Furthermore, there are still some uncertainties and lack of a full understanding of the virus as demonstrated in the high number new mutations arising worldwide and reinfections of already vaccinated individuals. To this end, efficient diagnostic tools and treatments are still urgently needed. In this context, the convergence of bioprinting and organ-on-a-chip technologies, either used alone or in combination, could possibly function as a prominent tool in addressing the current pandemic. This could enable facile advances of important tools, diagnostics, and better physiologically representative in vitro models specific to individuals allowing for faster and more accurate screening of therapeutics evaluating their efficacy and toxicity. This review will cover such technological advances and highlight what is needed for the field to mature for tackling the various needs for current and future pandemics as well as their relevancy towards precision medicine.
Collapse
Affiliation(s)
- Samson Afewerki
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Thiago Domingues Stocco
- Bioengineering Program, Technological and Scientific Institute, Brazil University, 08230-030, São Paulo, SP, Brazil; Faculty of Medical Sciences, Unicamp - State University of Campinas, 13083-877, Campinas, SP, Brazil
| | | | - André Sales Aguiar Furtado
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Gustavo Fernandes de Sousa
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Guillermo U Ruiz-Esparza
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA
| | - Thomas J Webster
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil; Hebei University of Technology, Tianjin, China
| | - Fernanda R Marciano
- Department of Physics, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Maria Strømme
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA.
| | - Anderson Oliveira Lobo
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil.
| |
Collapse
|
10
|
Jäkel AC, Heymann M, Simmel FC. Multiscale Biofabrication: Integrating Additive Manufacturing with DNA-Programmable Self-Assembly. Adv Biol (Weinh) 2023; 7:e2200195. [PMID: 36328598 DOI: 10.1002/adbi.202200195] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/23/2022] [Indexed: 11/06/2022]
Abstract
Structure and hierarchical organization are crucial elements of biological systems and are likely required when engineering synthetic biomaterials with life-like behavior. In this context, additive manufacturing techniques like bioprinting have become increasingly popular. However, 3D bioprinting, as well as other additive manufacturing techniques, show limited resolution, making it difficult to yield structures on the sub-cellular level. To be able to form macroscopic synthetic biological objects with structuring on this level, manufacturing techniques have to be used in conjunction with biomolecular nanotechnology. Here, a short overview of both topics and a survey of recent advances to combine additive manufacturing with microfabrication techniques and bottom-up self-assembly involving DNA, are given.
Collapse
Affiliation(s)
- Anna C Jäkel
- School of Natural Sciences, Department of Bioscience, Technical University Munich, Am Coulombwall 4a, 85748, Garching b. München, Germany
| | - Michael Heymann
- Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
| | - Friedrich C Simmel
- School of Natural Sciences, Department of Bioscience, Technical University Munich, Am Coulombwall 4a, 85748, Garching b. München, Germany
| |
Collapse
|
11
|
Cabral KA, Srivastava V, Graham AJ, Coyle MC, Stashko C, Weaver V, Gartner ZJ. Programming the Self-Organization of Endothelial Cells into Perfusable Microvasculature. Tissue Eng Part A 2023; 29:80-92. [PMID: 36181350 PMCID: PMC10266707 DOI: 10.1089/ten.tea.2022.0072] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/15/2022] [Indexed: 11/12/2022] Open
Abstract
The construction of three-dimensional (3D) microvascular networks with defined structures remains challenging. Emerging bioprinting strategies provide a means of patterning endothelial cells (ECs) into the geometry of 3D microvascular networks, but the microenvironmental cues necessary to promote their self-organization into cohesive and perfusable microvessels are not well known. To this end, we reconstituted microvessel formation in vitro by patterning thin lines of closely packed ECs fully embedded within a 3D extracellular matrix (ECM) and observed how different microenvironmental parameters influenced EC behaviors and their self-organization into microvessels. We found that the inclusion of fibrillar matrices, such as collagen I, into the ECM positively influenced cell condensation into extended geometries such as cords. We also identified the presence of a high-molecular-weight protein(s) in fetal bovine serum that negatively influenced EC condensation. This component destabilized cord structure by promoting cell protrusions and destabilizing cell-cell adhesions. Endothelial cords cultured in the presence of fibrillar collagen and in the absence of this protein activity were able to polarize, lumenize, incorporate mural cells, and support fluid flow. These optimized conditions allowed for the construction of branched and perfusable microvascular networks directly from patterned cells in as little as 3 days. These findings reveal important design principles for future microvascular engineering efforts based on bioprinting and micropatterning techniques. Impact statement Bioprinting is a potential strategy to achieve microvascularization in engineered tissues. However, the controlled self-organization of patterned endothelial cells into perfusable microvasculature remains challenging. We used DNA Programmed Assembly of Cells to create cell-dense, capillary-sized cords of endothelial cells with complete control over their structure. We optimized the matrix and media conditions to promote self-organization and maturation of these endothelial cords into stable and perfusable microvascular networks.
Collapse
Affiliation(s)
- Katelyn A. Cabral
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Austin J. Graham
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
| | - Maxwell C. Coyle
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, California, USA
| | - Connor Stashko
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Valerie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
- Center for Cellular Construction, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
12
|
Tripathi S, Mandal SS, Bauri S, Maiti P. 3D bioprinting and its innovative approach for biomedical applications. MedComm (Beijing) 2023; 4:e194. [PMID: 36582305 PMCID: PMC9790048 DOI: 10.1002/mco2.194] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 12/26/2022] Open
Abstract
3D bioprinting or additive manufacturing is an emerging innovative technology revolutionizing the field of biomedical applications by combining engineering, manufacturing, art, education, and medicine. This process involved incorporating the cells with biocompatible materials to design the required tissue or organ model in situ for various in vivo applications. Conventional 3D printing is involved in constructing the model without incorporating any living components, thereby limiting its use in several recent biological applications. However, this uses additional biological complexities, including material choice, cell types, and their growth and differentiation factors. This state-of-the-art technology consciously summarizes different methods used in bioprinting and their importance and setbacks. It also elaborates on the concept of bioinks and their utility. Biomedical applications such as cancer therapy, tissue engineering, bone regeneration, and wound healing involving 3D printing have gained much attention in recent years. This article aims to provide a comprehensive review of all the aspects associated with 3D bioprinting, from material selection, technology, and fabrication to applications in the biomedical fields. Attempts have been made to highlight each element in detail, along with the associated available reports from recent literature. This review focuses on providing a single platform for cancer and tissue engineering applications associated with 3D bioprinting in the biomedical field.
Collapse
Affiliation(s)
- Swikriti Tripathi
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Subham Shekhar Mandal
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Sudepta Bauri
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Pralay Maiti
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| |
Collapse
|
13
|
Wang B, Wang X, Kenneth A, Drena A, Pacheco A, Kalvin L, Ibrahim ES, Rossi PJ, Thatcher K, Lincoln J. Developing small-diameter vascular grafts with human amniotic membrane: long-term evaluation of transplantation outcomes in a small animal model. Biofabrication 2023; 15. [PMID: 36626826 DOI: 10.1088/1758-5090/acb1da] [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: 10/05/2022] [Accepted: 01/10/2023] [Indexed: 01/11/2023]
Abstract
While current clinical utilization of large vascular grafts for vascular transplantation is encouraging, tissue engineering of small grafts still faces numerous challenges. This study aims to investigate the feasibility of constructing a small vascular graft from decellularized amniotic membranes (DAMs). DAMs were rolled around a catheter and each of the resulting grafts was crosslinked with (a) 0.1% glutaraldehyde; (b) 1-ethyl-3-(3-dimethylaminopropyl) crbodiimidehydro-chloride (20 mM)-N-hydroxy-succinimide (10 mM); (c) 0.5% genipin; and (d) no-crosslinking, respectively. Our results demonstrated the feasibility of using a rolling technique followed by lyophilization to transform DAM into a vessel-like structure. The genipin-crosslinked DAM graft showed an improved integrated structure, prolonged stability, proper mechanical property, and superior biocompatibility. After transplantation in rat abdominal aorta, the genipin-crosslinked DAM graft remained patent up to 16 months, with both endothelial and smooth muscle cell regeneration, which suggests that the genipin-crosslinked DAM graft has great potential to beimplementedas a small tissue engineered graft for futurevasculartransplantation.
Collapse
Affiliation(s)
- Bo Wang
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Xiaolong Wang
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Allen Kenneth
- Biomedical Resource Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Alexander Drena
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States of America
| | - Arsenio Pacheco
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States of America
| | - Lindsey Kalvin
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Ei-Sayed Ibrahim
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Peter J Rossi
- Heart and Vascular Center, Froedtert Hospital, Milwaukee, WI 53226, United States of America
| | - Kaitlyn Thatcher
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Joy Lincoln
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| |
Collapse
|
14
|
Wang Z, Xiang L, Lin F, Tang Y, Cui W. 3D bioprinting of emulating homeostasis regulation for regenerative medicine applications. J Control Release 2023; 353:147-165. [PMID: 36423869 DOI: 10.1016/j.jconrel.2022.11.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022]
Abstract
Homeostasis is the most fundamental mechanism of physiological processes, occurring simultaneously as the production and outcomes of pathological procedures. Accompanied by manufacture and maturation of intricate and highly hierarchical architecture obtained from 3D bioprinting (three-dimension bioprinting), homeostasis has substantially determined the quality of printed tissues and organs. Instead of only shape imitation that has been the remarkable advances, fabrication for functionality to make artificial tissues and organs that act as real ones in vivo has been accepted as the optimized strategy in 3D bioprinting for the next several years. Herein, this review aims to provide not only an overview of 3D bioprinting, but also the main strategies used for homeostasis bioprinting. This paper briefly introduces the principles of 3D bioprinting system applied in homeostasis regulations firstly, and then summarizes the specific strategies and potential trend of homeostasis regulations using multiple types of stimuli-response biomaterials to maintain auto regulation, specifically displaying a brilliant prospect in hormone regulation of homeostasis with the most recently outbreak of vasculature fabrication. Finally, we discuss challenges and future prospects of homeostasis fabrication based on 3D bioprinting in regenerative medicine, hoping to further inspire the development of functional fabrication in 3D bioprinting.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Lei Xiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Feng Lin
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Yunkai Tang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.
| |
Collapse
|
15
|
Dell AC, Wagner G, Own J, Geibel JP. 3D Bioprinting Using Hydrogels: Cell Inks and Tissue Engineering Applications. Pharmaceutics 2022; 14:2596. [PMID: 36559090 PMCID: PMC9784738 DOI: 10.3390/pharmaceutics14122596] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022] Open
Abstract
3D bioprinting is transforming tissue engineering in medicine by providing novel methods that are precise and highly customizable to create biological tissues. The selection of a "cell ink", a printable formulation, is an integral part of adapting 3D bioprinting processes to allow for process optimization and customization related to the target tissue. Bioprinting hydrogels allows for tailorable material, physical, chemical, and biological properties of the cell ink and is suited for biomedical applications. Hydrogel-based cell ink formulations are a promising option for the variety of techniques with which bioprinting can be achieved. In this review, we will examine some of the current hydrogel-based cell inks used in bioprinting, as well as their use in current and proposed future bioprinting methods. We will highlight some of the biological applications and discuss the development of new hydrogels and methods that can incorporate the completed print into the tissue or organ of interest.
Collapse
Affiliation(s)
- Annika C. Dell
- The John B. Pierce Laboratory, Inc., New Haven, CT 06519, USA
- Fraunhofer IMTE, Fraunhofer Research Institution for Individualized and Cell-Based Medical Engineering, 23562 Lübeck, Germany
| | | | - Jason Own
- Yale University, New Haven, CT 06520, USA
| | - John P. Geibel
- The John B. Pierce Laboratory, Inc., New Haven, CT 06519, USA
- Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
16
|
Panda S, Hajra S, Mistewicz K, Nowacki B, In-Na P, Krushynska A, Mishra YK, Kim HJ. A focused review on three-dimensional bioprinting technology for artificial organ fabrication. Biomater Sci 2022; 10:5054-5080. [PMID: 35876134 DOI: 10.1039/d2bm00797e] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three-dimensional (3D) bioprinting technology has attracted a great deal of interest because it can be easily adapted to many industries and research sectors, such as biomedical, manufacturing, education, and engineering. Specifically, 3D bioprinting has provided significant advances in the medical industry, since such technology has led to significant breakthroughs in the synthesis of biomaterials, cells, and accompanying elements to produce composite living tissues. 3D bioprinting technology could lead to the immense capability of replacing damaged or injured tissues or organs with newly dispensed cell biomaterials and functional tissues. Several types of bioprinting technology and different bio-inks can be used to replicate cells and generate supporting units as complex 3D living tissues. Bioprinting techniques have undergone great advancements in the field of regenerative medicine to provide 3D printed models for numerous artificial organs and transplantable tissues. This review paper aims to provide an overview of 3D-bioprinting technologies by elucidating the current advancements, recent progress, opportunities, and applications in this field. It highlights the most recent advancements in 3D-bioprinting technology, particularly in the area of artificial organ development and cancer research. Additionally, the paper speculates on the future progress in 3D-bioprinting as a versatile foundation for several biomedical applications.
Collapse
Affiliation(s)
- Swati Panda
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea.
| | - Sugato Hajra
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea.
| | - Krystian Mistewicz
- Institute of Physics - Center for Science and Education, Silesian University of Technology, Krasińskiego 8, Katowice, Poland
| | - Bartłomiej Nowacki
- Faculty of Materials Engineering, Silesian University of Technology, Krasińskiego 8, Katowice, Poland
| | - Pichaya In-Na
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, 254 Phyathai Road, Wangmai, Pathumwan, Bangkok-10330, Thailand
| | - Anastasiia Krushynska
- Engineering and Technology Institute Groningen (ENTEG), Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, Netherlands
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, Alsion 2, 6400 Sønderborg, Denmark
| | - Hoe Joon Kim
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea. .,Robotics and Mechatronics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea
| |
Collapse
|
17
|
Nieto D, Jiménez G, Moroni L, López-Ruiz E, Gálvez-Martín P, Marchal JA. Biofabrication approaches and regulatory framework of metastatic tumor-on-a-chip models for precision oncology. Med Res Rev 2022; 42:1978-2001. [PMID: 35707911 PMCID: PMC9545141 DOI: 10.1002/med.21914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 12/14/2022]
Abstract
The complexity of the tumor microenvironment (TME) together with the development of the metastatic process are the main reasons for the failure of conventional anticancer treatment. In recent years, there is an increasing need to advance toward advanced in vitro models of cancer mimicking TME and simulating metastasis to understand the associated mechanisms that are still unknown, and to be able to develop personalized therapy. In this review, the commonly used alternatives and latest advances in biofabrication of tumor‐on‐chips, which allow the generation of the most sophisticated and optimized models for recapitulating the tumor process, are presented. In addition, the advances that have allowed these new models in the area of metastasis, cancer stem cells, and angiogenesis are summarized, as well as the recent integration of multiorgan‐on‐a‐chip systems to recapitulate natural metastasis and pharmacological screening against it. We also analyze, for the first time in the literature, the normative and regulatory framework in which these models could potentially be found, as well as the requirements and processes that must be fulfilled to be commercially implemented as in vitro study model. Moreover, we are focused on the possible regulatory pathways for their clinical application in precision medicine and decision making through the generation of personalized models with patient samples. In conclusion, this review highlights the synergistic combination of three‐dimensional bioprinting systems with the novel tumor/metastasis/multiorgan‐on‐a‐chip systems to generate models for both basic research and clinical applications to have devices useful for personalized oncology.
Collapse
Affiliation(s)
- Daniel Nieto
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, University of Maastricht, Universiteitssingel, Maastricht, The Netherlands.,Center for Biomedical Research (CIBM)/Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain
| | - Gema Jiménez
- Center for Biomedical Research (CIBM)/Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, University of Maastricht, Universiteitssingel, Maastricht, The Netherlands
| | - Elena López-Ruiz
- Center for Biomedical Research (CIBM)/Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain.,Department of Health Sciences, University of Jaén, Jaén, Spain
| | | | - Juan Antonio Marchal
- Center for Biomedical Research (CIBM)/Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| |
Collapse
|
18
|
Im H, Heo E, Song DH, Park J, Park H, Kang K, Chang JB. Fabrication of heterogeneous chemical patterns on stretchable hydrogels using single-photon lithography. SOFT MATTER 2022; 18:4402-4413. [PMID: 35635476 DOI: 10.1039/d2sm00253a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Curved hydrogel surfaces bearing chemical patterns are highly desirable in various applications, including artificial blood vessels, wearable electronics, and soft robotics. However, previous studies on the fabrication of chemical patterns on hydrogels employed two-photon lithography, which is still not widely accessible to most laboratories. This work demonstrates a new patterning technique for fabricating curved hydrogels with chemical patterns on their surfaces without two-photon microscopy. In this work, we show that exposing hydrogels in fluorophore solutions to single photons via confocal microscopy enables the patterning of fluorophores on hydrogels. By applying this technique to highly stretchable hydrogels, we demonstrate three applications: (1) improving pattern resolution by fabricating patterns on stretched hydrogels and then returning the hydrogels to their initial, unstretched length; (2) modifying the local stretchability of hydrogels at a microscale resolution; and (3) fabricating perfusable microchannels with chemical patterns by winding chemically patterned hydrogels around a template, embedding the hydrogels in a second hydrogel, and then removing the template. The patterning method demonstrated in this work may facilitate a better mimicking of the physicochemical properties of organs in tissue engineering and may be used to make hydrogel robots with specific chemical functionalities.
Collapse
Affiliation(s)
- Haeseong Im
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| | - Eunseok Heo
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| | - Dae-Hyeon Song
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| | - Jeongwon Park
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| | - Hyeonbin Park
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
- Division of Advanced Materials, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Kibum Kang
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| | - Jae-Byum Chang
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
19
|
Smart Bioinks for the Printing of Human Tissue Models. Biomolecules 2022; 12:biom12010141. [PMID: 35053289 PMCID: PMC8773823 DOI: 10.3390/biom12010141] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/05/2022] [Accepted: 01/13/2022] [Indexed: 12/11/2022] Open
Abstract
3D bioprinting has tremendous potential to revolutionize the field of regenerative medicine by automating the process of tissue engineering. A significant number of new and advanced bioprinting technologies have been developed in recent years, enabling the generation of increasingly accurate models of human tissues both in the healthy and diseased state. Accordingly, this technology has generated a demand for smart bioinks that can enable the rapid and efficient generation of human bioprinted tissues that accurately recapitulate the properties of the same tissue found in vivo. Here, we define smart bioinks as those that provide controlled release of factors in response to stimuli or combine multiple materials to yield novel properties for the bioprinting of human tissues. This perspective piece reviews the existing literature and examines the potential for the incorporation of micro and nanotechnologies into bioinks to enhance their properties. It also discusses avenues for future work in this cutting-edge field.
Collapse
|
20
|
Liu H, Yang X, Cheng X, Zhao G, Zheng G, Li X, Dong R. Theoretical and Experimental Research on Multi-Layer Vessel-like Structure Printing Based on 3D Bio-Printing Technology. MICROMACHINES 2021; 12:mi12121517. [PMID: 34945366 PMCID: PMC8709219 DOI: 10.3390/mi12121517] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 11/23/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Traditional autologous transplantation has become a severe issue due to insufficient donors. Artificial blood vessel is an effective method for the treatment of major vascular diseases, such as heart and peripheral blood vessel diseases. However, the traditional single-material printing technology has been unable to meet the users’ demand for product functional complexity, which is not only reflected in the field of industrial manufacturing, but also in the field of functional vessel-like structure regeneration. In order to achieve the printing and forming of multi-layer vessel-like structures, this paper carries out theoretical and experimental research on the printing and forming of a multi-layer vessel-like structure based on multi-material 3D bioprinting technology. Firstly, theoretical analysis has been explored to research the relationship among the different parameters in the process of vessel forming, and further confirm the synchronous relationship among the extrusion rate of material, the tangential speed of the rotating rod, and the movement speed of the platform. Secondly, sodium alginate and gelatin have been used as the experimental materials to manufacture the vessel-like structure, and the corrected parameter of the theoretical analysis is further verified. Finally, the cell-loaded materials have been printed and analyzed, and cell viability is more than 90%, which provides support for the research of multi-layer vessel-like structure printing.
Collapse
|
21
|
Hackethal J, Weihs AM, Karner L, Metzger M, Dungel P, Hennerbichler S, Redl H, Teuschl-Woller AH. Novel Human Placenta-Based Extract for Vascularization Strategies in Tissue Engineering. Tissue Eng Part C Methods 2021; 27:616-632. [PMID: 34714165 DOI: 10.1089/ten.tec.2021.0173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
There is critical unmet need for new vascularized tissues to support or replace injured tissues and organs. Various synthetic and natural materials were already established for use of two-dimensional (2D) and three-dimensional (3D) in vitro neovascularization assays, however, they still cannot mimic the complex functions of the sum of the extracellular matrix (ECM) in native intact tissue. Currently, this issue is only addressed by artificial products such as Matrigel™, which comprises a complex mixture of ECM proteins, extracted from animal tumor tissue. Despite its outstanding bioactivity, the isolation from tumor tissue hinders its translation into clinical applications. Since nonhuman ECM proteins may cause immune reactions, as are frequently observed in clinical trials, human ECM proteins represent the best option when aiming for clinical applications. Here, we describe an effective method of isolating a human placenta substrate (hpS) that induces the spontaneous formation of an interconnected network of green fluorescence-labeled human umbilical vein endothelial cells (gfpHUVECs) in vitro. The substrate was biochemically characterized by using a combination of bicinchoninic acid (BCA) assay, DNA, and glycosaminoglycan (GAG) content assays, sodium dodecyl sulfate/polyacrylamide gel electrophoresis (SDS-PAGE) analysis and Western blot, angiogenesis arrays, chromatographic thrombin detection, high performance liquid chromatography (HPLC)-based amino acid quantification analysis, and assessment of antimicrobial properties. 2D in vitro cell culture experiments have been performed to determine the vasculogenic potential of hpS, which demonstrated that cell networks developed on hpS show a significantly higher degree of complexity (number of tubules/junctions; total/mean tube length) when compared with Matrigel. As 3D cell culture techniques represent a more accurate representation of the in vivo condition, the substrate was 3D solidified using various natural polymers. 3D in vitro vasculogenesis assays have been performed by seeding gfpHUVECs in an hpS-fibrinogen clot. In conclusion, hpS provides a potent human/material-based alternative to xenogenic-material-based biomaterials for vascularization strategies in tissue engineering.
Collapse
Affiliation(s)
- Johannes Hackethal
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Trauma Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Anna Maria Weihs
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
| | - Lisa Karner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Trauma Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Magdalena Metzger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Trauma Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Peter Dungel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Trauma Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Simone Hennerbichler
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Trauma Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andreas Herbert Teuschl-Woller
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
| |
Collapse
|
22
|
Gao G, Ahn M, Cho WW, Kim BS, Cho DW. 3D Printing of Pharmaceutical Application: Drug Screening and Drug Delivery. Pharmaceutics 2021; 13:1373. [PMID: 34575448 PMCID: PMC8465948 DOI: 10.3390/pharmaceutics13091373] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/20/2021] [Accepted: 08/29/2021] [Indexed: 12/22/2022] Open
Abstract
Advances in three-dimensional (3D) printing techniques and the development of tailored biomaterials have facilitated the precise fabrication of biological components and complex 3D geometrics over the past few decades. Moreover, the notable growth of 3D printing has facilitated pharmaceutical applications, enabling the development of customized drug screening and drug delivery systems for individual patients, breaking away from conventional approaches that primarily rely on transgenic animal experiments and mass production. This review provides an extensive overview of 3D printing research applied to drug screening and drug delivery systems that represent pharmaceutical applications. We classify several elements required by each application for advanced pharmaceutical techniques and briefly describe state-of-the-art 3D printing technology consisting of cells, bioinks, and printing strategies that satisfy requirements. Furthermore, we discuss the limitations of traditional approaches by providing concrete examples of drug screening (organoid, organ-on-a-chip, and tissue/organ equivalent) and drug delivery systems (oral/vaginal/rectal and transdermal/surgical drug delivery), followed by the introduction of recent pharmaceutical investigations using 3D printing-based strategies to overcome these challenges.
Collapse
Affiliation(s)
- Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China;
| | - Minjun Ahn
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| | - Won-Woo Cho
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| | - Byoung-Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Kyungbuk, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| |
Collapse
|
23
|
Fazal F, Raghav S, Callanan A, Koutsos V, Radacsi N. Recent advancements in the bioprinting of vascular grafts. Biofabrication 2021; 13. [PMID: 34102613 DOI: 10.1088/1758-5090/ac0963] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023]
Abstract
Recent advancements in the bioinks and three-dimensional (3D) bioprinting methods used to fabricate vascular constructs are summarized herein. Critical biomechanical properties required to fabricate an ideal vascular graft are highlighted, as well as various testing methods have been outlined to evaluate the bio-fabricated grafts as per the Food and Drug Administration (FDA) and International Organization for Standardization (ISO) guidelines. Occlusive artery disease and cardiovascular disease are the major causes of death globally. These diseases are caused by the blockage in the arteries, which results in a decreased blood flow to the tissues of major organs in the body, such as the heart. Bypass surgery is often performed using a vascular graft to re-route the blood flow. Autologous grafts represent a gold standard for such bypass surgeries; however, these grafts may be unavailable due to the previous harvesting or possess a poor quality. Synthetic grafts serve well for medium to large-sized vessels, but they fail when used to replace small-diameter vessels, generally smaller than 6 mm. Various tissue engineering approaches have been used to address the urgent need for vascular graft that can withstand hemodynamic blood pressure and has the ability to grow and remodel. Among these approaches, 3D bioprinting offers an attractive solution to construct patient-specific vessel grafts with layered biomimetic structures.
Collapse
Affiliation(s)
- Faraz Fazal
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, EH9 3FB Edinburgh, United Kingdom.,Department of Mechanical Engineering, University of Engineering and Technology, Lahore, (New Campus) Pakistan
| | - Sakshika Raghav
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, EH9 3FB Edinburgh, United Kingdom
| | - Anthony Callanan
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, The King's Buildings, EH9 3JL Edinburgh, United Kingdom
| | - Vasileios Koutsos
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, EH9 3FB Edinburgh, United Kingdom
| | - Norbert Radacsi
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, EH9 3FB Edinburgh, United Kingdom
| |
Collapse
|
24
|
3D Bioprinting for fabrication of tissue models of COVID-19 infection. Essays Biochem 2021; 65:503-518. [PMID: 34028514 DOI: 10.1042/ebc20200129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/19/2022]
Abstract
Over the last few decades, the world has witnessed multiple viral pandemics, the current severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) pandemic being the worst and most devastating one, claiming millions of lives worldwide. Physicians, scientists, and engineers worldwide have joined hands in dealing with the current situation at an impressive speed and efficiency. One of the major reasons for the delay in response is our limited understanding of the mechanism of action and individual effects of the virus on different tissues and organs. Advances in 3D bioprinting have opened up a whole new area to explore and utilize the technology in fabricating models of these tissues and organs, recapitulating in vivo environment. These biomimetic models can not only be utilized in learning the infection pathways and drug toxicology studies but also minimize the need for animal models and shorten the time span for human clinical trials. The current review aims to integrate the existing developments in bioprinting techniques, and their implementation to develop tissue models, which has implications for SARS-CoV-2 infection. Future translation of these models has also been discussed with respect to the pandemic.
Collapse
|
25
|
Zarket BC, Wang H, Subraveti SN, Raghavan SR. Multilayer tubes that constrict, dilate, and curl in response to stimuli. SOFT MATTER 2021; 17:4180-4190. [PMID: 33881039 DOI: 10.1039/d0sm01704c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Tubular structures in nature have the ability to respond to their environment-for example, blood vessels can constrict or dilate, thereby regulating flow velocity and blood pressure. These tubes have multiple concentric layers, with each layer having a distinct composition and properties. Inspired by such natural structures, we have synthesized responsive multilayer tubes in the laboratory without resorting to complex equipment such as a 3-D printer. Each layer of our tubes is a polymer gel formed by free-radical polymerization of water-soluble monomers. We can precisely control the inner diameter of the tube, the number of layers in the tube wall, and the thickness and chemistry of each layer. Tubes synthesized in this manner are robust, flexible, and stretchable. Moreover, our technique allows us to incorporate stimuli-responsive polymers into distinct regions of these tubes, and the resulting tubes can change their shape in response to external stimuli such as pH or temperature. In the case of laterally patterned tubes, the tube can be made to constrict or dilate over a particular segment-a behavior that is reminiscent of blood vessels. In the case of longitudinally patterned tubes, a straight tube can be induced to systematically curl into a coil. The versatility of our technique is further shown by constructing complex tubular architectures, including branched networks. On the whole, the polymeric tubes shown in this paper exhibit remarkable properties that cannot be realized by other techniques. Such tubes could find utility in biomedical engineering to construct anatomically realistic mimics of various tissues.
Collapse
Affiliation(s)
- Brady C Zarket
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, USA.
| | - Hanchu Wang
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, USA.
| | - Sai N Subraveti
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, USA.
| | - Srinivasa R Raghavan
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, USA.
| |
Collapse
|
26
|
Augustine R, Kalva SN, Ahmad R, Zahid AA, Hasan S, Nayeem A, McClements L, Hasan A. 3D Bioprinted cancer models: Revolutionizing personalized cancer therapy. Transl Oncol 2021; 14:101015. [PMID: 33493799 PMCID: PMC7823217 DOI: 10.1016/j.tranon.2021.101015] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
After cardiovascular disease, cancer is the leading cause of death worldwide with devastating health and economic consequences, particularly in developing countries. Inter-patient variations in anti-cancer drug responses further limit the success of therapeutic interventions. Therefore, personalized medicines approach is key for this patient group involving molecular and genetic screening and appropriate stratification of patients to treatment regimen that they will respond to. However, the knowledge related to adequate risk stratification methods identifying patients who will respond to specific anti-cancer agents is still lacking in many cancer types. Recent advancements in three-dimensional (3D) bioprinting technology, have been extensively used to generate representative bioengineered tumor in vitro models, which recapitulate the human tumor tissues and microenvironment for high-throughput drug screening. Bioprinting process involves the precise deposition of multiple layers of different cell types in combination with biomaterials capable of generating 3D bioengineered tissues based on a computer-aided design. Bioprinted cancer models containing patient-derived cancer and stromal cells together with genetic material, extracellular matrix proteins and growth factors, represent a promising approach for personalized cancer therapy screening. Both natural and synthetic biopolymers have been utilized to support the proliferation of cells and biological material within the personalized tumor models/implants. These models can provide a physiologically pertinent cell-cell and cell-matrix interactions by mimicking the 3D heterogeneity of real tumors. Here, we reviewed the potential applications of 3D bioprinted tumor constructs as personalized in vitro models in anticancer drug screening and in the establishment of precision treatment regimens.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| | - Sumama Nuthana Kalva
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Rashid Ahmad
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Alap Ali Zahid
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Shajia Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Ajisha Nayeem
- Department of Biotechnology, St. Mary's College, Thrissur, 680020, Kerala, India
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, 2007, NSW, Australia
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| |
Collapse
|
27
|
Biofabrication in Congenital Cardiac Surgery: A Plea from the Operating Theatre, Promise from Science. MICROMACHINES 2021; 12:mi12030332. [PMID: 33800971 PMCID: PMC8004062 DOI: 10.3390/mi12030332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/08/2021] [Accepted: 03/18/2021] [Indexed: 12/11/2022]
Abstract
Despite significant advances in numerous fields of biofabrication, clinical application of biomaterials combined with bioactive molecules and/or cells largely remains a promise in an individualized patient settings. Three-dimensional (3D) printing and bioprinting evolved as promising techniques used for tissue-engineering, so that several kinds of tissue can now be printed in layers or as defined structures for replacement and/or reconstruction in regenerative medicine and surgery. Besides technological, practical, ethical and legal challenges to solve, there is also a gap between the research labs and the patients' bedside. Congenital and pediatric cardiac surgery mostly deal with reconstructive patient-scenarios when defects are closed, various segments of the heart are connected, valves are implanted. Currently available biomaterials lack the potential of growth and conduits, valves derange over time surrendering patients to reoperations. Availability of viable, growing biomaterials could cancel reoperations that could entail significant public health benefit and improved quality-of-life. Congenital cardiac surgery is uniquely suited for closing the gap in translational research, rapid application of new techniques, and collaboration between interdisciplinary teams. This article provides a succinct review of the state-of-the art clinical practice and biofabrication strategies used in congenital and pediatric cardiac surgery, and highlights the need and avenues for translational research and collaboration.
Collapse
|
28
|
Serex L, Sharma K, Rizov V, Bertsch A, McKinney JD, Renaud P. Microfluidic-assisted bioprinting of tissues and organoids at high cell concentrations. Biofabrication 2021; 13. [DOI: 10.1088/1758-5090/abca80] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
|
29
|
Çelebi-Saltik B, Öteyaka MÖ, Gökçinar-Yagci B. Stem cell-based small-diameter vascular grafts in dynamic culture. Connect Tissue Res 2021; 62:151-163. [PMID: 31379220 DOI: 10.1080/03008207.2019.1651848] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: Transplantation of autologous and/or allogeneic blood vessels is the most convenient treatment for vascular diseases. With regard to extensive need for blood vessels, developments in vascular tissue engineering are contributing greatly. In this study, our aim is to create intact small-diameter tubular vascular grafts cultivated in pulsatile flow bioreactor. Materials and Methods: CD146+ cell-based small-diameter vascular grafts were fabricated with ECM/glycosaminoglycans and polyurethane nanofibers. Characterization of the vascular graft was performed by SEM and WST-1. To mimic blood circulation in the bioreactor, human CD34+ cells cultured in megakaryocytes/platelets medium; then these cells were transferred inside of the vascular graft to mimic blood circulation. Cell differentiation was evaluated by flow cytometry and colony assay. Wright-Giemsa staining and polyploidy analysis were performed to show the differentiated cell population inside of the vascular graft. Anti-thrombogenic properties of the blood vessel were demonstrated by IF. Results: Polyurethane nanofibers provided a suitable environment for Human umbilical cord vein endothelial cells (HUVECs), and no significant cytotoxic effect was observed. Scanning electron microscopy (SEM) analysis of the tubular graft showed that under perfusion HUVECs, smooth muscle cells (SMCs) and fibroblasts formed layers that aligned on each other, respectively. The vascular graft was strong with a tensile strength of 0.70 MPa and elastic modulus of 0.007 GPa. When cultured in a bioreactor system, platelet adhesion to the vascular graft was remarkably low. Conclusion: In conclusion, this vascular graft may hold the potential to regenerate functional small-diameter vessels for cardiovascular tissue repair.
Collapse
Affiliation(s)
- Betül Çelebi-Saltik
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University , Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University , Ankara, Turkey
| | - Mustafa Özgür Öteyaka
- Mechatronic Program, Eskişehir Vocational School, Eskişehir Osmangazi University , Eskişehir, Turkey
| | - Beyza Gökçinar-Yagci
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University , Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University , Ankara, Turkey
| |
Collapse
|
30
|
Ando M, Ikeguchi R, Aoyama T, Tanaka M, Noguchi T, Miyazaki Y, Akieda S, Nakayama K, Matsuda S. Long-Term Outcome of Sciatic Nerve Regeneration Using Bio3D Conduit Fabricated from Human Fibroblasts in a Rat Sciatic Nerve Model. Cell Transplant 2021; 30:9636897211021357. [PMID: 34105391 PMCID: PMC8193652 DOI: 10.1177/09636897211021357] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 11/17/2022] Open
Abstract
Previously, we developed a Bio3D conduit fabricated from human fibroblasts and reported a significantly better outcome compared with artificial nerve conduit in the treatment of rat sciatic nerve defect. The purpose of this study is to investigate the long-term safety and nerve regeneration of Bio3D conduit compared with treatments using artificial nerve conduit and autologous nerve transplantation.We used 15 immunodeficient rats and randomly divided them into three groups treated with Bio3D (n = 5) conduit, silicon tube (n = 5), and autologous nerve transplantation (n = 5). We developed Bio3D conduits composed of human fibroblasts and bridged the 5 mm nerve gap created in the rat sciatic nerve. The same procedures were performed to bridge the 5 mm gap with a silicon tube. In the autologous nerve group, we removed the 5 mm sciatic nerve segment and transplanted it. We evaluated the nerve regeneration 24 weeks after surgery.Toe dragging was significantly better in the Bio3D group (0.20 ± 0.28) than in the silicon group (0.6 ± 0.24). The wet muscle weight ratios of the tibial anterior muscle of the Bio3D group (79.85% ± 5.47%) and the autologous nerve group (81.74% ± 2.83%) were significantly higher than that of the silicon group (66.99% ± 3.51%). The number of myelinated axons and mean myelinated axon diameter was significantly higher in the Bio3D group (14708 ± 302 and 5.52 ± 0.44 μm) and the autologous nerve group (14927 ± 5089 and 6.04 ± 0.85 μm) than the silicon group (7429 ± 1465 and 4.36 ± 0.21 μm). No tumors were observed in any of the rats in the Bio3D group at 24 weeks after surgery.The Bio3D group showed significantly better nerve regeneration and there was no significant difference between the Bio3D group and the nerve autograft group in all endpoints.
Collapse
Affiliation(s)
| | - Ryosuke Ikeguchi
- Ryosuke Ikeguchi, Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Morrison KA, Weinreb RH, Dong X, Toyoda Y, Jin JL, Bender R, Mukherjee S, Spector JA. Facilitated self-assembly of a prevascularized dermal/epidermal collagen scaffold. Regen Med 2020; 15:2273-2283. [PMID: 33325258 DOI: 10.2217/rme-2020-0070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Introduction: Resurfacing complex full thickness wounds requires free tissue transfer which creates donor site morbidity. We describe a method to fabricate a skin flap equivalent with a hierarchical microvascular network. Materials & methods: We fabricated a flap of skin-like tissue containing a hierarchical vascular network by sacrificing Pluronic® F127 macrofibers and interwoven microfibers within collagen encapsulating human pericytes and fibroblasts. Channels were seeded with smooth muscle and endothelial cells. Constructs were topically seeded with keratinocytes. Results: After 28 days in culture, multiphoton microscopy revealed a hierarchical interconnected network of macro- and micro-vessels; larger vessels (>100 μm) were lined with a monolayer endothelial neointima and a subendothelial smooth muscle neomedia. Neoangiogenic sprouts formed in the collagen protodermis and pericytes self-assembled around both fabricated vessels and neoangiogenic sprouts. Conclusion: We fabricated a prevascularized scaffold containing a hierarchical 3D network of interconnected macro- and microchannels within a collagen protodermis subjacent to an overlying protoepidermis with the potential for recipient microvascular anastomosis.
Collapse
Affiliation(s)
- Kerry A Morrison
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA.,Plastic Surgery Resident Physician affiliated with the Hansjorg Wyss Department of Plastic Surgery, New York University Langone Medical Center, New York, NY 10016, USA
| | - Ross H Weinreb
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Xue Dong
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Yoshiko Toyoda
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA.,Plastic Surgery Resident Physician affiliated with the Division of Plastic Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julia L Jin
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Ryan Bender
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Sushmita Mukherjee
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 14850, USA
| | - Jason A Spector
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA.,Nancy E. & Peter C. Meinig School of Bioengineering, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
32
|
Abstract
Microvasculature functions at the tissue and cell level, regulating local mass exchange of oxygen and nutrient-rich blood. While there has been considerable success in the biofabrication of large- and small-vessel replacements, functional microvasculature has been particularly challenging to engineer due to its size and complexity. Recently, three-dimensional bioprinting has expanded the possibilities of fabricating sophisticated microvascular systems by enabling precise spatiotemporal placement of cells and biomaterials based on computer-aided design. However, there are still significant challenges facing the development of printable biomaterials that promote robust formation and controlled 3D organization of microvascular networks. This review provides a thorough examination and critical evaluation of contemporary biomaterials and their specific roles in bioprinting microvasculature. We first provide an overview of bioprinting methods and techniques that enable the fabrication of microvessels. We then offer an in-depth critical analysis on the use of hydrogel bioinks for printing microvascularized constructs within the framework of current bioprinting modalities. We end with a review of recent applications of bioprinted microvasculature for disease modeling, drug testing, and tissue engineering, and conclude with an outlook on the challenges facing the evolution of biomaterials design for bioprinting microvasculature with physiological complexity.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael Yost
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
33
|
Richard C, Neild A, Cadarso VJ. The emerging role of microfluidics in multi-material 3D bioprinting. LAB ON A CHIP 2020; 20:2044-2056. [PMID: 32459222 DOI: 10.1039/c9lc01184f] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
To assist the transition of 3D bioprinting technology from simple lab-based tissue fabrication, to fully functional and implantable organs, the technology must not only provide shape control, but also functional control. This can be accomplished by replicating the cellular composition of the native tissue at the microscale, such that cell types interact to provide the desired function. There is therefore a need for precise, controllable, multi-material printing that could allow for high, possibly even single cell, resolution. This paper aims to draw attention to technological advancements made in 3D bioprinting that target the lack of multi-material, and/or multi cell-type, printing capabilities of most current devices. Unlike other reviews in the field, which largely focus on variations in single-material 3D bioprinting involving the standard methods of extrusion-based, droplet-based, laser-based, or stereolithographic methods; this review concentrates on sophisticated multi-material 3D bioprinting using multi-cartridge printheads, co-axial nozzles and microfluidic-enhanced printing nozzles.
Collapse
Affiliation(s)
- Cynthia Richard
- Laboratory for Micro Systems, Department of Mechanical and Aerospace Engineering, Monash University, Clayton, VIC 3800, Australia.
| | | | | |
Collapse
|
34
|
Pakhomova C, Popov D, Maltsev E, Akhatov I, Pasko A. Software for Bioprinting. Int J Bioprint 2020; 6:279. [PMID: 33088988 PMCID: PMC7557344 DOI: 10.18063/ijb.v6i3.279] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
The bioprinting of heterogeneous organs is a crucial issue. To reach the complexity of such organs, there is a need for highly specialized software that will meet all requirements such as accuracy, complexity, and others. The primary objective of this review is to consider various software tools that are used in bioprinting and to reveal their capabilities. The sub-objective was to consider different approaches for the model creation using these software tools. Related articles on this topic were analyzed. Software tools are classified based on control tools, general computer-aided design (CAD) tools, tools to convert medical data to CAD formats, and a few highly specialized research-project tools. Different geometry representations are considered, and their advantages and disadvantages are considered applicable to heterogeneous volume modeling and bioprinting. The primary factor for the analysis is suitability of the software for heterogeneous volume modeling and bioprinting or multimaterial three-dimensional printing due to the commonality of these technologies. A shortage of specialized suitable software tools is revealed. There is a need to develop a new application area such as computer science for bioprinting which can contribute significantly in future research work.
Collapse
Affiliation(s)
- Catherine Pakhomova
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow.,Institute of Engineering Physics for Biomedicine, NRNU Mephi, Moscow
| | - Dmitry Popov
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow
| | - Eugenii Maltsev
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow
| | - Iskander Akhatov
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow
| | - Alexander Pasko
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow.,The National Centre for Computer Animation, Bournemouth University, UK
| |
Collapse
|
35
|
Colle J, Blondeel P, De Bruyne A, Bochar S, Tytgat L, Vercruysse C, Van Vlierberghe S, Dubruel P, Declercq H. Bioprinting predifferentiated adipose-derived mesenchymal stem cell spheroids with methacrylated gelatin ink for adipose tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:36. [PMID: 32206922 DOI: 10.1007/s10856-020-06374-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 03/03/2020] [Indexed: 06/10/2023]
Abstract
The increasing number of mastectomies results in a greater demand for breast reconstruction characterized by simplicity and a low complication profile. Reconstructive surgeons are investigating tissue engineering (TE) strategies to overcome the current surgical drawbacks. 3D bioprinting is the rising technique for the fabrication of large tissue constructs which provides a potential solution for unmet clinical needs in breast reconstruction building on decades of experience in autologous fat grafting, adipose-derived mesenchymal stem cell (ASC) biology and TE. A scaffold was bioprinted using encapsulated ASC spheroids in methacrylated gelatin ink (GelMA). Uniform ASC spheroids with an ideal geometry and diameter for bioprinting were formed, using a high-throughput non-adhesive agarose microwell system. ASC spheroids in adipogenic differentiation medium (ADM) were evaluated through live/dead staining, histology (HE, Oil Red O), TEM and RT-qPCR. Viable spheroids were obtained for up to 14 days post-printing and showed multilocular microvacuoles and successful differentiation toward mature adipocytes shown by gene expression analysis. Moreover, spheroids were able to assemble at random in GelMA, creating a macrotissue. Combining the advantage of microtissues to self-assemble and the controlled organization by bioprinting technologies, these ASC spheroids can be useful as building blocks for the engineering of soft tissue implants.
Collapse
Affiliation(s)
- Julien Colle
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium.
- Department of Plastic Surgery, University Hospital Gent, Gent, Belgium.
| | - Phillip Blondeel
- Department of Plastic Surgery, University Hospital Gent, Gent, Belgium
| | - Axelle De Bruyne
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium
| | - Silke Bochar
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium
| | - Liesbeth Tytgat
- Polymer Chemistry and Biomaterials Group, Department of Organic Chemistry, Faculty of Sciences, Gent University, Krijgslaan 281, 9000, Gent, Belgium
| | - Chris Vercruysse
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Department of Organic Chemistry, Faculty of Sciences, Gent University, Krijgslaan 281, 9000, Gent, Belgium
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group, Department of Organic Chemistry, Faculty of Sciences, Gent University, Krijgslaan 281, 9000, Gent, Belgium
| | - Heidi Declercq
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium
| |
Collapse
|
36
|
Mohamed MGA, Ambhorkar P, Samanipour R, Yang A, Ghafoor A, Kim K. Microfluidics-based fabrication of cell-laden microgels. BIOMICROFLUIDICS 2020; 14:021501. [PMID: 32161630 PMCID: PMC7058428 DOI: 10.1063/1.5134060] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/16/2020] [Indexed: 05/02/2023]
Abstract
Microfluidic principles have been extensively utilized as powerful tools to fabricate controlled monodisperse cell-laden hydrogel microdroplets for various biological applications, especially tissue engineering. In this review, we report recent advances in microfluidic-based droplet fabrication and provide our rationale to justify the superiority of microfluidics-based techniques over other microtechnology methods in achieving the encapsulation of cells within hydrogels. The three main components of such a system-hydrogels, cells, and device configurations-are examined thoroughly. First, the characteristics of various types of hydrogels including natural and synthetic types, especially concerning cell encapsulation, are examined. This is followed by the elucidation of the reasoning behind choosing specific cells for encapsulation. Next, in addition to a detailed discussion of their respective droplet formation mechanisms, various device configurations including T-junctions, flow-focusing, and co-flowing that aid in achieving cell encapsulation are critically reviewed. We then present an outlook on the current applications of cell-laden hydrogel droplets in tissue engineering such as 3D cell culturing, rapid generation and repair of tissues, and their usage as platforms for studying cell-cell and cell-microenvironment interactions. Finally, we shed some light upon the prospects of microfluidics-based production of cell-laden microgels and propose some directions for forthcoming research that can aid in overcoming challenges currently impeding the translation of the technology into clinical success.
Collapse
Affiliation(s)
- Mohamed G. A. Mohamed
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Pranav Ambhorkar
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Roya Samanipour
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Annie Yang
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Ali Ghafoor
- Irving K. Barber School of Arts and Sciences, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | | |
Collapse
|
37
|
Li L, Qin S, Peng J, Chen A, Nie Y, Liu T, Song K. Engineering gelatin-based alginate/carbon nanotubes blend bioink for direct 3D printing of vessel constructs. Int J Biol Macromol 2020; 145:262-271. [DOI: 10.1016/j.ijbiomac.2019.12.174] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/03/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022]
|
38
|
Baltazar T, Merola J, Catarino C, Xie CB, Kirkiles-Smith NC, Lee V, Hotta S, Dai G, Xu X, Ferreira FC, Saltzman WM, Pober JS, Karande P. Three Dimensional Bioprinting of a Vascularized and Perfusable Skin Graft Using Human Keratinocytes, Fibroblasts, Pericytes, and Endothelial Cells. Tissue Eng Part A 2019; 26:227-238. [PMID: 31672103 DOI: 10.1089/ten.tea.2019.0201] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Multilayered skin substitutes comprising allogeneic cells have been tested for the treatment of nonhealing cutaneous ulcers. However, such nonnative skin grafts fail to permanently engraft because they lack dermal vascular networks important for integration with the host tissue. In this study, we describe the fabrication of an implantable multilayered vascularized bioengineered skin graft using 3D bioprinting. The graft is formed using one bioink containing human foreskin dermal fibroblasts (FBs), human endothelial cells (ECs) derived from cord blood human endothelial colony-forming cells (HECFCs), and human placental pericytes (PCs) suspended in rat tail type I collagen to form a dermis followed by printing with a second bioink containing human foreskin keratinocytes (KCs) to form an epidermis. In vitro, KCs replicate and mature to form a multilayered barrier, while the ECs and PCs self-assemble into interconnected microvascular networks. The PCs in the dermal bioink associate with EC-lined vascular structures and appear to improve KC maturation. When these 3D printed grafts are implanted on the dorsum of immunodeficient mice, the human EC-lined structures inosculate with mouse microvessels arising from the wound bed and become perfused within 4 weeks after implantation. The presence of PCs in the printed dermis enhances the invasion of the graft by host microvessels and the formation of an epidermal rete. Impact Statement Three Dimensional printing can be used to generate multilayered vascularized human skin grafts that can potentially overcome the limitations of graft survival observed in current avascular skin substitutes. Inclusion of human pericytes in the dermal bioink appears to improve both dermal and epidermal maturation.
Collapse
Affiliation(s)
- Tânia Baltazar
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
| | - Jonathan Merola
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - Carolina Catarino
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York.,Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Catherine B Xie
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
| | | | - Vivian Lee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Stephanie Hotta
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Frederico C Ferreira
- Department of Bioengineering and Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Jordan S Pober
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
| | - Pankaj Karande
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York.,Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
39
|
Liu Y, Yang Y, Shen Y. Tubular Microcapsules with Polysaccharide Membranes Based on a Co-axial Microfluidic Chip. ACS Biomater Sci Eng 2019; 5:6281-6289. [DOI: 10.1021/acsbiomaterials.9b01077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yanting Liu
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Yuanyuan Yang
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Yajing Shen
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen 518055, China
| |
Collapse
|
40
|
|
41
|
|
42
|
Three-dimensional bioprinting for organ bioengineering: promise and pitfalls. Curr Opin Organ Transplant 2019; 23:649-656. [PMID: 30234736 DOI: 10.1097/mot.0000000000000581] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Loss of organ function is a critical issue that threatens a patient's life. Currently, the only available treatment is organ transplantation; however, shortage of donor organs, histocompatibility, and life-long immunosuppression present major challenges. Three-dimensional bioprinting technology holds a promising solution for treating organ failure by fabricating autologous tissues and organs for transplantation. To biofabricate a functional tissue, target-cell types are combined with an appropriate biomaterial for structural support and a bioink that supports cell function and maturation. Bioprinted structures can mimic the native tissue shape and functionality. RECENT FINDINGS The main goal of three-dimensional bioprinting is to produce functional tissues/organs; however, whole organ printing has not been achieved. There have been recent advances in the successful three-dimensional bioprinting of numerous tissues. This review will discuss the types of bioprinters, biomaterials, bioinks, and the fabrication of various constructs for repair of vascular, cartilage, skin, cardiac, and liver tissues. These bioprinted tissue constructs have the potential to be used to treat tissues and organs that have been damaged by injury or disease. SUMMARY Three-dimensional bioprinting technology offers the ability to fabricate three-dimensional tissue structures with high precision, fidelity, and stability at human clinical scale. The creation of complex tissue architectures with heterogeneous compositions has the potential to revolutionize transplantation of tissues and organs.
Collapse
|
43
|
Sheiko SS, Dobrynin AV. Architectural Code for Rubber Elasticity: From Supersoft to Superfirm Materials. Macromolecules 2019. [DOI: 10.1021/acs.macromol.9b01127] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Sergei S. Sheiko
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-3290, United States
| | - Andrey V. Dobrynin
- Department of Polymer Science, University of Akron, Akron, Ohio 44325-3909, United States
| |
Collapse
|
44
|
Abdollahi S, Boktor J, Hibino N. Bioprinting of freestanding vascular grafts and the regulatory considerations for additively manufactured vascular prostheses. Transl Res 2019; 211:123-138. [PMID: 31201778 PMCID: PMC6702084 DOI: 10.1016/j.trsl.2019.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/15/2019] [Accepted: 05/23/2019] [Indexed: 12/31/2022]
Abstract
Vasculature is the network of blood vessels of an organ or body part that allow for the exchange of nutrients and waste to and from every cell, thus establishing a circulatory equilibrium. Vascular health is at risk from a variety of conditions that includes disease and trauma. In some cases, medical therapy can alleviate the impacts of the condition. Intervention is needed in other instances to restore the health of abnormal vasculature. The main approaches to treat vascular conditions are endovascular procedures and open vascular reconstruction that often requires a graft to accomplish. However, current vascular prostheses have limitations that include size mismatch with the native vessel, risk of immunogenicity from allografts and xenografts, and unavailability of autografts. In this review, we discuss efforts in bioprinting, an emerging method for vascular reconstruction. This includes an overview of 3D printing processes and materials, graft characterization strategies and the regulatory aspects to consider for the commercialization of 3D bioprinted vascular prostheses.
Collapse
Affiliation(s)
- Sara Abdollahi
- Division of Cardiac Surgery, Johns Hopkins Hospital, Baltimore, Maryland
| | - Joseph Boktor
- Division of Cardiac Surgery, Johns Hopkins Hospital, Baltimore, Maryland; Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - Narutoshi Hibino
- Division of Cardiac Surgery, Johns Hopkins Hospital, Baltimore, Maryland.
| |
Collapse
|
45
|
Unagolla JM, Jayasuriya AC. Enhanced cell functions on graphene oxide incorporated 3D printed polycaprolactone scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 102:1-11. [PMID: 31146979 PMCID: PMC6546300 DOI: 10.1016/j.msec.2019.04.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 03/28/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023]
Abstract
For tissue engineering applications, a porous scaffold with an interconnected network is essential to facilitate the cell attachment and proliferation in a three dimensional (3D) structure. This study aimed to fabricate the scaffolds by an extrusion-based 3D printer using a blend of polycaprolactone (PCL), and graphene oxide (GO) as a favorable platform for bone tissue engineering. The mechanical properties, morphology, biocompatibility, and biological activities such as cell proliferation and differentiation were studied concerning the two different pore sizes; 400 μm, and 800 μm, and also with two different GO content; 0.1% (w/w) and 0.5% (w/w). The compressive strength of the scaffolds was not significantly changed due to the small amount of GO, but, as expected scaffolds with 400 μm pores showed a higher compressive modulus in comparison to the scaffolds with 800 μm pores. The data indicated that the cell attachment and proliferation were increased by adding a small amount of GO. According to the results, pore size did not play a significant role in cell proliferation and differentiation. Alkaline Phosphate (ALP) activity assay further confirmed that the GO increase the ALP activity and further Elemental analysis of Calcium and Phosphorous showed that the GO increased the mineralization compared to PCL only scaffolds. Western blot analysis showed the porous structure facilitate the secretion of bone morphogenic protein-2 (BMP-2) and osteopontin at both day 7 and 14 which galvanizes the osteogenic capability of PCL and PCL + GO scaffolds.
Collapse
Affiliation(s)
- Janitha M Unagolla
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43607, USA
| | - Ambalangodage C Jayasuriya
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43607, USA; Department of Orthopedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA.
| |
Collapse
|
46
|
A versatile perfusion bioreactor and endothelializable photo cross-linked tubes of gelatin methacryloyl as promising tools in tissue engineering. ACTA ACUST UNITED AC 2019; 64:397-406. [DOI: 10.1515/bmt-2018-0015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022]
Abstract
Abstract
Size and function of bioartificial tissue models are still limited due to the lack of blood vessels and dynamic perfusion for nutrient supply. In this study, we evaluated the use of cytocompatible methacryl-modified gelatin for the fabrication of a hydrogel-based tube by dip-coating and subsequent photo-initiated cross-linking. The wall thickness of the tubes and the diameter were tuned by the degree of gelatin methacryl-modification and the number of dipping cycles. The dipping temperature of the gelatin solution was adjusted to achieve low viscous fluids of approximately 0.1 Pa s and was different for gelatin derivatives with different modification degrees. A versatile perfusion bioreactor for the supply of surrounding tissue models was developed, which can be adapted to several geometries and sizes of blood-vessel mimicking tubes. The manufactured bendable gelatin tubes were permeable for water and dissolved substances, like Nile Blue and serum albumin. As a proof of concept, human fibroblasts in a three-dimensional collagen tissue model were successfully supplied with nutrients via the central gelatin tube under dynamic conditions for 2 days. Moreover, the tubes could be used as scaffolds to build-up a functional and viable endothelial layer. Hence, the presented tools can contribute to solving current challenges in tissue engineering.
Collapse
|
47
|
Li S, Wang K, Hu Q, Zhang C, Wang B. Direct-write and sacrifice-based techniques for vasculatures. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109936. [PMID: 31500055 DOI: 10.1016/j.msec.2019.109936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 05/22/2019] [Accepted: 07/01/2019] [Indexed: 12/27/2022]
Abstract
Fabricating biomimetic vasculatures is considered one of the greatest challenges in tissue regeneration due to their complex structures across various length scales. Many strategies have been investigated on how to fabricate tissue-engineering vasculatures (TEVs), including vascular-like and vascularized structures that can replace their native counterparts. The advancement of additive manufacturing (AM) technologies has enabled a wide range of fabrication techniques that can directly-write TEVs with complex and delicate structures. Meanwhile, sacrifice-based techniques, which rely on the removal of encapsulated sacrificial templates to form desired cavity-like structures, have also been widely studied. This review will specifically focus on the two most promising methods in these recently developed technologies, which are the direct-write method and the sacrifice-based method. The performance, advantages, and shortcomings of each technique are analyzed and compared. In the discussion, we list current challenges in this field and present our vision of next-generation TEVs technologies. Perspectives on future research in this field are given at the end.
Collapse
Affiliation(s)
- Shuai Li
- Georgia Tech Manufacturing Institute, Georgia Institute of Technology, Atlanta, GA 30332, USA; Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Kan Wang
- Georgia Tech Manufacturing Institute, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Qingxi Hu
- Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China; Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, Shanghai University, Shanghai 200072, China; National Demonstration Center for Experimental Engineering Training Education, Shanghai University, Shanghai 200444, China.
| | - Chuck Zhang
- Georgia Tech Manufacturing Institute, Georgia Institute of Technology, Atlanta, GA 30332, USA; H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ben Wang
- Georgia Tech Manufacturing Institute, Georgia Institute of Technology, Atlanta, GA 30332, USA; H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
48
|
Eswaramoorthy SD, Ramakrishna S, Rath SN. Recent advances in three-dimensional bioprinting of stem cells. J Tissue Eng Regen Med 2019; 13:908-924. [PMID: 30866145 DOI: 10.1002/term.2839] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 02/01/2019] [Accepted: 02/21/2019] [Indexed: 12/29/2022]
Abstract
In spite of being a new field, three-dimensional (3D) bioprinting has undergone rapid growth in the recent years. Bioprinting methods offer a unique opportunity for stem cell distribution, positioning, and differentiation at the microscale to make the differentiated architecture of any tissue while maintaining precision and control over the cellular microenvironment. Bioprinting introduces a wide array of approaches to modify stem cell fate. This review discusses these methodologies of 3D bioprinting stem cells. Fabricating a fully operational tissue or organ construct with a long life will be the most significant challenge of 3D bioprinting. Once this is achieved, a whole human organ can be fabricated for the defect place at the site of surgery.
Collapse
Affiliation(s)
- Sindhuja D Eswaramoorthy
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad (IITH), Sangareddy, Telangana, India
| | - Seeram Ramakrishna
- Centre for Nanofibers & Nanotechnology, NUS Nanoscience & Nanotechnology Initiative, Singapore
| | - Subha N Rath
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad (IITH), Sangareddy, Telangana, India
| |
Collapse
|
49
|
Papaioannou TG, Manolesou D, Dimakakos E, Tsoucalas G, Vavuranakis M, Tousoulis D. 3D Bioprinting Methods and Techniques: Applications on Artificial Blood Vessel Fabrication. ACTA CARDIOLOGICA SINICA 2019; 35:284-289. [PMID: 31249458 PMCID: PMC6533576 DOI: 10.6515/acs.201905_35(3).20181115a] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 11/15/2018] [Indexed: 12/16/2022]
Abstract
Three-dimensional (3D) printing has rapidly evolved, with major applications in the field of medicine. One of the greatest advances leading to 3D bioprinting was the development of biomaterials, cells and supporting components for the fabrication of functional living tissues. Several different methods and techniques of 3D bioprinting are briefly described in this review article, and applications of 3D printing for the fabrication of artificial blood vessels and grafts are presented. Advances in additive manufacturing techniques, medical imaging modalities, biomaterials and cellular engineering will lead to further developments in the fabrication of patient-specific vascular tissue constructs. Future multidisciplinary research and innovations are expected to further transform the fields of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
| | - Danae Manolesou
- Biomedical Engineering Unit, First Department of Cardiology, Medical School
| | | | - Gregory Tsoucalas
- Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Manolis Vavuranakis
- Third Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Athens
| | | |
Collapse
|
50
|
Qasim M, Haq F, Kang MH, Kim JH. 3D printing approaches for cardiac tissue engineering and role of immune modulation in tissue regeneration. Int J Nanomedicine 2019; 14:1311-1333. [PMID: 30863063 PMCID: PMC6388753 DOI: 10.2147/ijn.s189587] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Conventional tissue engineering, cell therapy, and current medical approaches were shown to be successful in reducing mortality rate and complications caused by cardiovascular diseases (CVDs). But still they have many limitations to fully manage CVDs due to complex composition of native myocardium and microvascularization. Fabrication of fully functional construct to replace infarcted area or regeneration of progenitor cells is important to address CVDs burden. Three-dimensional (3D) printed scaffolds and 3D bioprinting technique have potential to develop fully functional heart construct that can integrate with native tissues rapidly. In this review, we presented an overview of 3D printed approaches for cardiac tissue engineering, and advances in 3D bioprinting of cardiac construct and models. We also discussed role of immune modulation to promote tissue regeneration.
Collapse
Affiliation(s)
- Muhammad Qasim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, South Korea,
| | - Farhan Haq
- Department of Biosciences, Comsats University, Islamabad, Pakistan
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, South Korea,
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, South Korea,
| |
Collapse
|