451
|
Unleashing bioprinting technology through patent intelligence. Drug Discov Today 2021; 26:1547-1555. [PMID: 33571470 DOI: 10.1016/j.drudis.2021.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/01/2020] [Accepted: 02/02/2021] [Indexed: 10/22/2022]
Abstract
Bioprinting technology is here to revolutionize the healthcare sector by providing customized solutions to the medicine industry. A review of the bioprinting patent documents provides insight into the development of this technology. A patent search was conducted using open-access patent databases and the raw data were refined by manually checking and eliminating irrelevant patents followed by hand-coding with master keywords. Graphics created from the data set facilitate the generation of patent intelligence on current bioprinting technologies in terms of prominent patent-filing countries and institutions, territorial biases in the areas of technology, and applications in the drug discovery process.
Collapse
|
452
|
Ruihan X, Weijie B, Zhihai W, Yaohong W. Start-up stage with improved resolution for an electric field-assisted fused deposition. RSC Adv 2021; 11:7397-7404. [PMID: 35423235 PMCID: PMC8694952 DOI: 10.1039/d0ra07795j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/29/2021] [Indexed: 11/29/2022] Open
Abstract
Electric field-assisted fused deposition modeling (E-FDM) is a promising technique in the field of 3D printing. This paper studies the start-up stage of the printing, which is a process of liquid gradually deforming and making an initial contact with the substrate under the action of electric stress. Polycaprolactone, a popular material for biomedicine, is selected as the printing material. With a home-built E-FDM system, the nozzle-to-substrate distance and the nozzle and substrate temperatures are all held steady. With a photography system, the process of meniscus deformation is recorded. And by image processing methods, the meniscus length and the volume of liquid at the nozzle can be obtained. At a set of initial liquid volumes (V i), nozzle voltage is ramped to a fixed value at a fixed rate. The effects of V i on the meniscus deformation during the start-up stage of the printing are examined. For sufficiently small V i, the meniscus deforms into a conical (Taylor cone) shape, and a fine jet with a diameter much smaller than the nozzle diameter appears. For sufficiently large V i, the meniscus exhibits a spindle shape when it touches the substrate. At an intermediate V i, a Taylor cone is formed, tending to eject a fine jet. After a short period of stagnation or even a slight retraction, no liquid is emitted. Through this study, it is suggested that for high-resolution printing, ramping the voltage at small V i may be preferable. This proposition is preliminarily confirmed in a direct writing test.
Collapse
Affiliation(s)
- Xu Ruihan
- Faculty of Information, Beijing University of Technology Beijing 100124 China
- Key Laboratory of Optoelectronics Technology, Beijing University of Technology Beijing 100124 China
| | - Bao Weijie
- Faculty of Information, Beijing University of Technology Beijing 100124 China
- Key Laboratory of Optoelectronics Technology, Beijing University of Technology Beijing 100124 China
| | - Wang Zhihai
- Faculty of Information, Beijing University of Technology Beijing 100124 China
- Key Laboratory of Optoelectronics Technology, Beijing University of Technology Beijing 100124 China
| | - Wang Yaohong
- Center for Applied Mathematics, Tianjin University Tianjin 300072 China
| |
Collapse
|
453
|
Recent progress in induced pluripotent stem cell-derived 3D cultures for cardiac regeneration. Cell Tissue Res 2021; 384:231-240. [PMID: 33544212 DOI: 10.1007/s00441-021-03414-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/10/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are the leading cause of death in the world due to the high incidence of the diseases coupled with the limited therapeutic options. In recent years, advances in regenerative medicine have emerged as a promising treatment. Differentiation of induced pluripotent stem cells (iPSCs) into cardiac cells and emerging technologies allowing arrangement of cells into complex 3D tissue-like structures open new frontiers for transplantation and engraftment of these tissue patches onto the damaged heart. Despite the cells integrating and presenting initial neovascularization, the functional and electric properties of these patches are still not comparable with those of the host cardiac tissue. Future research optimizing maturation and integration of the iPSC-derived cardiomyocytes is paramount for cardiac cell therapy to attain clinical use. Herein, we will review the state of the art and the different approaches to constructing these 3D transplantable structures.
Collapse
|
454
|
Agarwal T, Onesto V, Lamboni L, Ansari A, Maiti TK, Makvandi P, Vosough M, Yang G. Engineering biomimetic intestinal topological features in 3D tissue models: retrospects and prospects. Biodes Manuf 2021. [DOI: 10.1007/s42242-020-00120-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
455
|
Gao J, Ding X, Yu X, Chen X, Zhang X, Cui S, Shi J, Chen J, Yu L, Chen S, Ding J. Cell-Free Bilayered Porous Scaffolds for Osteochondral Regeneration Fabricated by Continuous 3D-Printing Using Nascent Physical Hydrogel as Ink. Adv Healthc Mater 2021; 10:e2001404. [PMID: 33225617 DOI: 10.1002/adhm.202001404] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/23/2020] [Indexed: 12/12/2022]
Abstract
Cartilage is difficult to self-repair and it is more challenging to repair an osteochondral defects concerning both cartilage and subchondral bone. Herein, it is hypothesized that a bilayered porous scaffold composed of a biomimetic gelatin hydrogel may, despite no external seeding cells, induce osteochondral regeneration in vivo after being implanted into mammal joints. This idea is confirmed based on the successful continuous 3D-printing of the bilayered scaffolds combined with the sol-gel transition of the aqueous solution of a gelatin derivative (physical gelation) and photocrosslinking of the gelatin methacryloyl (gelMA) macromonomers (chemical gelation). At the direct printing step, a nascent physical hydrogel is extruded, taking advantage of non-Newtonian and thermoresponsive rheological properties of this 3D-printing ink. In particular, a series of crosslinked gelMA (GelMA) and GelMA-hydroxyapatite bilayered hydrogel scaffolds are fabricated to evaluate the influence of the spacing of 3D-printed filaments on osteochondral regeneration in a rabbit model. The moderately spaced scaffolds output excellent regeneration of cartilage with cartilaginous lacunae and formation of subchondral bone. Thus, tricky rheological behaviors of soft matter can be employed to improve 3D-printing, and the bilayered hybrid scaffold resulting from the continuous 3D-printing is promising as a biomaterial to regenerate articular cartilage.
Collapse
Affiliation(s)
- Jingming Gao
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| | - Xiaoquan Ding
- Center of Sports Medicine Department of Sports Medicine Huashan Hospital and State Key Laboratory of Molecular Engineering of Polymers Fudan University Shanghai 200040 China
| | - Xiaoye Yu
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| | - Xiaobin Chen
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| | - Xingyu Zhang
- Center of Sports Medicine Department of Sports Medicine Huashan Hospital and State Key Laboratory of Molecular Engineering of Polymers Fudan University Shanghai 200040 China
| | - Shuquan Cui
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| | - Jiayue Shi
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| | - Jun Chen
- Center of Sports Medicine Department of Sports Medicine Huashan Hospital and State Key Laboratory of Molecular Engineering of Polymers Fudan University Shanghai 200040 China
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| | - Shiyi Chen
- Center of Sports Medicine Department of Sports Medicine Huashan Hospital and State Key Laboratory of Molecular Engineering of Polymers Fudan University Shanghai 200040 China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| |
Collapse
|
456
|
Sümbelli Y, Emir Diltemiz S, Say MG, Ünlüer ÖB, Ersöz A, Say R. In situ and non-cytotoxic cross-linking strategy for 3D printable biomaterials. SOFT MATTER 2021; 17:1008-1015. [PMID: 33284939 DOI: 10.1039/d0sm01734e] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
3D bioprinting allows the production of patient-specific tissue constructs with desired structural characteristics such as high resolution, controlled swelling degree, and controlled degradation behavior by mostly using hydrogels. Crosslinking of hydrogels is an essential parameter in bioprinting applications, which is beneficial for tuning structural specifications. In this study, gelatin-alginate-whey protein isolate based hydrogels have been used for 3D printing structures in a layer-by-layer fashion. These structures were cross-linked by the Amino Acid (monomer) Decorated and Light Underpinning Conjugation Approach (ANADOLUCA) method, which is a unique, non-invasive photosensitive cross-linking technique for protein-based mixtures. In that aim, hydrogel properties (e.g., printability, biocompatibility, rheologic and mechanical behavior) and cross-linking properties (e.g., swelling and degradation behavior) were studied. Results were compared with UV and ionic cross-linking techniques, which are the abundantly used techniques in such studies. The results showed that the ANADOLUCA method can be used for in situ cross-linking under mild conditions for the printing of bio-inks, and the proposed method can be used as an alternative for UV-based and chemical cross-linking techniques.
Collapse
Affiliation(s)
- Yiğitcan Sümbelli
- Chemistry Department, Faculty of Science, Eskişehir Technical University, 26470 Eskisehir, Turkey.
| | | | | | | | | | | |
Collapse
|
457
|
Pennarossa G, Arcuri S, De Iorio T, Gandolfi F, Brevini TAL. Current Advances in 3D Tissue and Organ Reconstruction. Int J Mol Sci 2021; 22:E830. [PMID: 33467648 PMCID: PMC7830719 DOI: 10.3390/ijms22020830] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Bi-dimensional culture systems have represented the most used method to study cell biology outside the body for over a century. Although they convey useful information, such systems may lose tissue-specific architecture, biomechanical effectors, and biochemical cues deriving from the native extracellular matrix, with significant alterations in several cellular functions and processes. Notably, the introduction of three-dimensional (3D) platforms that are able to re-create in vitro the structures of the native tissue, have overcome some of these issues, since they better mimic the in vivo milieu and reduce the gap between the cell culture ambient and the tissue environment. 3D culture systems are currently used in a broad range of studies, from cancer and stem cell biology, to drug testing and discovery. Here, we describe the mechanisms used by cells to perceive and respond to biomechanical cues and the main signaling pathways involved. We provide an overall perspective of the most recent 3D technologies. Given the breadth of the subject, we concentrate on the use of hydrogels, bioreactors, 3D printing and bioprinting, nanofiber-based scaffolds, and preparation of a decellularized bio-matrix. In addition, we report the possibility to combine the use of 3D cultures with functionalized nanoparticles to obtain highly predictive in vitro models for use in the nanomedicine field.
Collapse
Affiliation(s)
- Georgia Pennarossa
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Sharon Arcuri
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Teresina De Iorio
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy;
| | - Tiziana A. L. Brevini
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| |
Collapse
|
458
|
Khan A, Alamry KA, Asiri AM. Multifunctional Biopolymers‐Based Composite Materials for Biomedical Applications: A Systematic Review. ChemistrySelect 2021. [DOI: 10.1002/slct.202003978] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ajahar Khan
- Faculty of Science Department of Chemistry King Abdulaziz University Jeddah 21589 Saudi Arabia
| | - Khalid A. Alamry
- Faculty of Science Department of Chemistry King Abdulaziz University Jeddah 21589 Saudi Arabia
| | - Abdullah M. Asiri
- Faculty of Science Department of Chemistry King Abdulaziz University Jeddah 21589 Saudi Arabia
- Centre of Excellence for Advanced Materials Research King Abdulaziz University Jeddah 21589 Saudi Arabia
| |
Collapse
|
459
|
Daly AC, Prendergast ME, Hughes AJ, Burdick JA. Bioprinting for the Biologist. Cell 2021; 184:18-32. [PMID: 33417859 DOI: 10.1016/j.cell.2020.12.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/29/2020] [Accepted: 12/01/2020] [Indexed: 12/30/2022]
Abstract
Building tissues from scratch to explore entirely new cell configurations could revolutionize fundamental understanding in biology. Bioprinting is an emerging technology to do this. Although typically applied to engineer tissues for therapeutic tissue repair or drug screening, there are many opportunities for bioprinting within biology, such as for exploring cellular crosstalk or cellular morphogenesis. The overall goals of this Primer are to provide an overview of bioprinting with the biologist in mind, outline the steps in extrusion bioprinting (the most widely used and accessible technology), and discuss alternative bioprinting technologies and future opportunities for bioprinting in biology.
Collapse
Affiliation(s)
- Andrew C Daly
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Alex J Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
460
|
Simon-Yarza T, Labour MN, Aid R, Letourneur D. Channeled polysaccharide-based hydrogel reveals influence of curvature to guide endothelial cell arrangement in vessel-like structures. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 118:111369. [DOI: 10.1016/j.msec.2020.111369] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023]
|
461
|
Vanaei S, Parizi M, Vanaei S, Salemizadehparizi F, Vanaei H. An Overview on Materials and Techniques in 3D Bioprinting Toward Biomedical Application. ENGINEERED REGENERATION 2021. [DOI: 10.1016/j.engreg.2020.12.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
462
|
|
463
|
Sekar MP, Budharaju H, Zennifer A, Sethuraman S, Vermeulen N, Sundaramurthi D, Kalaskar DM. Current standards and ethical landscape of engineered tissues-3D bioprinting perspective. J Tissue Eng 2021; 12:20417314211027677. [PMID: 34377431 PMCID: PMC8330463 DOI: 10.1177/20417314211027677] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/08/2021] [Indexed: 01/17/2023] Open
Abstract
Tissue engineering is an evolving multi-disciplinary field with cutting-edge technologies and innovative scientific perceptions that promise functional regeneration of damaged tissues/organs. Tissue engineered medical products (TEMPs) are biomaterial-cell products or a cell-drug combination which is injected, implanted or topically applied in the course of a therapeutic or diagnostic procedure. Current tissue engineering strategies aim at 3D printing/bioprinting that uses cells and polymers to construct living tissues/organs in a layer-by-layer fashion with high 3D precision. However, unlike conventional drugs or therapeutics, TEMPs and 3D bioprinted tissues are novel therapeutics and need different regulatory protocols for clinical trials and commercialization processes. Therefore, it is essential to understand the complexity of raw materials, cellular components, and manufacturing procedures to establish standards that can help to translate these products from bench to bedside. These complexities are reflected in the regulations and standards that are globally in practice to prevent any compromise or undue risks to patients. This review comprehensively describes the current legislations, standards for TEMPs with a special emphasis on 3D bioprinted tissues. Based on these overviews, challenges in the clinical translation of TEMPs & 3D bioprinted tissues/organs along with their ethical concerns and future perspectives are discussed.
Collapse
Affiliation(s)
- Muthu Parkkavi Sekar
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Allen Zennifer
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Niki Vermeulen
- Department of Science, Technology and Innovation Studies, School of Social and Political Science, University of Edinburgh, High School Yards, Edinburgh, UK
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | | |
Collapse
|
464
|
Askari M, Afzali Naniz M, Kouhi M, Saberi A, Zolfagharian A, Bodaghi M. Recent progress in extrusion 3D bioprinting of hydrogel biomaterials for tissue regeneration: a comprehensive review with focus on advanced fabrication techniques. Biomater Sci 2021; 9:535-573. [DOI: 10.1039/d0bm00973c] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Over the last decade, 3D bioprinting has received immense attention from research communities to bridge the divergence between artificially engineered tissue constructs and native tissues.
Collapse
Affiliation(s)
- Mohsen Askari
- Department of Engineering
- School of Science and Technology
- Nottingham Trent University
- Nottingham NG11 8NS
- UK
| | - Moqaddaseh Afzali Naniz
- Department of Engineering
- School of Science and Technology
- Nottingham Trent University
- Nottingham NG11 8NS
- UK
| | - Monireh Kouhi
- Biomaterials Research Group
- Department of Materials Engineering
- Isfahan University of Technology
- Isfahan
- Iran
| | - Azadeh Saberi
- Nanotechnology and Advanced Materials Department
- Materials and Energy Research Center
- Tehran
- Iran
| | | | - Mahdi Bodaghi
- Department of Engineering
- School of Science and Technology
- Nottingham Trent University
- Nottingham NG11 8NS
- UK
| |
Collapse
|
465
|
Wang Z, Kapadia W, Li C, Lin F, Pereira RF, Granja PL, Sarmento B, Cui W. Tissue-specific engineering: 3D bioprinting in regenerative medicine. J Control Release 2021; 329:237-256. [DOI: 10.1016/j.jconrel.2020.11.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022]
|
466
|
Weng T, Zhang W, Xia Y, Wu P, Yang M, Jin R, Xia S, Wang J, You C, Han C, Wang X. 3D bioprinting for skin tissue engineering: Current status and perspectives. J Tissue Eng 2021; 12:20417314211028574. [PMID: 34345398 PMCID: PMC8283073 DOI: 10.1177/20417314211028574] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/10/2021] [Indexed: 12/25/2022] Open
Abstract
Skin and skin appendages are vulnerable to injury, requiring rapidly reliable regeneration methods. In recent years, 3D bioprinting has shown potential for wound repair and regeneration. 3D bioprinting can be customized for skin shape with cells and other materials distributed precisely, achieving rapid and reliable production of bionic skin substitutes, therefore, meeting clinical and industrial requirements. Additionally, it has excellent performance with high resolution, flexibility, reproducibility, and high throughput, showing great potential for the fabrication of tissue-engineered skin. This review introduces the common techniques of 3D bioprinting and their application in skin tissue engineering, focusing on the latest research progress in skin appendages (hair follicles and sweat glands) and vascularization, and summarizes current challenges and future development of 3D skin printing.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Zhang
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yilan Xia
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pan Wu
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| | - Min Yang
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| | - Ronghua Jin
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| | - Sizhan Xia
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| | - Jialiang Wang
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| | - Chuangang You
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| | - Chunmao Han
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xingang Wang
- Department of Burns & Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Trauma and Burns of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
467
|
Seyedsalehi A, Daneshmandi L, Barajaa M, Riordan J, Laurencin CT. Fabrication and characterization of mechanically competent 3D printed polycaprolactone-reduced graphene oxide scaffolds. Sci Rep 2020; 10:22210. [PMID: 33335152 PMCID: PMC7747749 DOI: 10.1038/s41598-020-78977-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022] Open
Abstract
The ability to produce constructs with a high control over the bulk geometry and internal architecture has situated 3D printing as an attractive fabrication technique for scaffolds. Various designs and inks are actively investigated to prepare scaffolds for different tissues. In this work, we prepared 3D printed composite scaffolds comprising polycaprolactone (PCL) and various amounts of reduced graphene oxide (rGO) at 0.5, 1, and 3 wt.%. We employed a two-step fabrication process to ensure an even mixture and distribution of the rGO sheets within the PCL matrix. The inks were prepared by creating composite PCL-rGO films through solvent evaporation casting that were subsequently fed into the 3D printer for extrusion. The resultant scaffolds were seamlessly integrated, and 3D printed with high fidelity and consistency across all groups. This, together with the homogeneous dispersion of the rGO sheets within the polymer matrix, significantly improved the compressive strength and stiffness by 185% and 150%, respectively, at 0.5 wt.% rGO inclusion. The in vitro response of the scaffolds was assessed using human adipose-derived stem cells. All scaffolds were cytocompatible and supported cell growth and viability. These mechanically reinforced and biologically compatible 3D printed PCL-rGO scaffolds are a promising platform for regenerative engineering applications.
Collapse
Affiliation(s)
- Amir Seyedsalehi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, 293 Farmington Avenue, Farmington, CT, 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA
| | - Leila Daneshmandi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, 293 Farmington Avenue, Farmington, CT, 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA
| | - Mohammed Barajaa
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, 293 Farmington Avenue, Farmington, CT, 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA
| | - John Riordan
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, 293 Farmington Avenue, Farmington, CT, 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, 293 Farmington Avenue, Farmington, CT, 06030, USA.
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA.
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA.
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
468
|
Ma H, Ma Z, Chen Q, Li W, Liu X, Ma X, Mao Y, Yang H, Ma H, Wang J. Bifunctional, Copper-Doped, Mesoporous Silica Nanosphere-Modified, Bioceramic Scaffolds for Bone Tumor Therapy. Front Chem 2020; 8:610232. [PMID: 33363114 PMCID: PMC7755992 DOI: 10.3389/fchem.2020.610232] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 10/26/2020] [Indexed: 12/11/2022] Open
Abstract
In the traditional surgical intervention procedure, residual tumor cells may potentially cause tumor recurrence. In addition, large bone defects caused by surgery are difficult to self-repair. Thus, it is necessary to design a bioactive scaffold that can not only kill residual tumor cells but also promote bone defect regeneration simultaneously. Here, we successfully developed Cu-containing mesoporous silica nanosphere-modified β-tricalcium phosphate (Cu-MSN-TCP) scaffolds, with uniform and dense nanolayers with spherical morphology via 3D printing and spin coating. The scaffolds exhibited coating time- and laser power density-dependent photothermal performance, which favored the effective killing of tumor cells under near-infrared laser irradiation. Furthermore, the prepared scaffolds favored the proliferation and attachment of rabbit bone marrow-derived mesenchymal stem cells and stimulated the gene expression of osteogenic markers. Overall, Cu-MSN-TCP scaffolds can be considered for complete eradication of residual bone tumor cells and simultaneous healing of large bone defects, which may provide a novel and effective strategy for bone tumor therapy. In the future, such Cu-MSN-TCP scaffolds may function as carriers of anti-cancer drugs or immune checkpoint inhibitors in chemo-/photothermal or immune-/photothermal therapy of bone tumors, favoring for effective treatment.
Collapse
Affiliation(s)
- Hongshi Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qufei Chen
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangfei Liu
- Department of Orthopaedic Surgery, Shanghai Zhongye Hospital, Shanghai, China
| | - Xiaojun Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanqing Mao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
469
|
Lian M, Han Y, Sun B, Xu L, Wang X, Ni B, Jiang W, Qiao Z, Dai K, Zhang X. A multifunctional electrowritten bi-layered scaffold for guided bone regeneration. Acta Biomater 2020; 118:83-99. [PMID: 32853801 DOI: 10.1016/j.actbio.2020.08.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/21/2020] [Accepted: 08/18/2020] [Indexed: 12/22/2022]
Abstract
The guided bone regeneration (GBR) concept has been extensively utilized to treat maxillofacial bone defects in clinical practice. However, the repair efficacy of currently available GBR membranes is often compromised by their limited bone regeneration potential and deficient antibacterial activity. In this study, inspired by the bi-layered structure design of the commonly used Bio-GideⓇmembrane, we designed and fabricated a new kind of multifunctional bi-layered "GBR scaffold" combining solution electrospinning writing (SEW) and solution electrospinning (SES) techniques using a single SEW printer. Copper-loaded mesoporous silica nanoparticles (Cu@MSNs) were incorporated into the poly(lactic-co-glycolic acid)/gelatin (PLGA/Gel, denoted as PG) fiber matrix to construct a composite PG-Cu@MSNs fibrous scaffold. The obtained GBR scaffold consisted of a loose and porous SEW layer to support and facilitate bone ingrowth, and a dense and compact SES layer to resist non-osteoblast interference. The resulting enhanced mechanical properties, coordinated degradation profile, and facile preparation procedure imparted the composite scaffold with good clinical feasibility. In vitro biological experiments indicate that the PG-Cu@MSNs composite scaffold exhibited favorable osteogenic and antibacterial properties. Furthermore, an in vivo rat periodontal defect model further confirmed the promising bone regeneration efficacy of the PG-Cu@MSNs scaffold. In conclusion, the developed electrowritten Cu@MSNs-incorporated bi-layered scaffold with hierarchical architecture and concurrent osteogenic and antibacterial functions may hold great potential for application in GBR.
Collapse
Affiliation(s)
- Meifei Lian
- Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yu Han
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong university School of Medicine, Shanghai 200125, China
| | - Binbin Sun
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong university School of Medicine, Shanghai 200125, China
| | - Ling Xu
- Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaofeng Wang
- School of Environmental Science, Nanjing Xiaozhuang University, Excellent Science and Technology Innovation Group of Jiangsu Province, Nanjing, 211171, China
| | - Bing Ni
- School of Life Science, East China Normal University, Shanghai 200241, China
| | - Wenbo Jiang
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong university School of Medicine, Shanghai 200125, China
| | - Zhiguang Qiao
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong university School of Medicine, Shanghai 200125, China; Department of Orthopaedic Surgery, Renji Hospital, South Campus, Shanghai Jiao Tong University School of Medicine, Shanghai 201112, China.
| | - Kerong Dai
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong university School of Medicine, Shanghai 200125, China.
| | - Xiuyin Zhang
- Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
470
|
Chakraborty PK, Azadmanjiri J, Pavithra CLP, Wang X, Masood SH, Dey SR, Wang J. Advancements in Therapeutics via 3D Printed Multifunctional Architectures from Dispersed 2D Nanomaterial Inks. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004900. [PMID: 33185035 DOI: 10.1002/smll.202004900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/01/2020] [Indexed: 06/11/2023]
Abstract
2D nanomaterials (2DNMs) possess fascinating properties and are found in multifarious devices and applications including energy storage devices, new generation of battery technologies, sensor devices, and more recently in biomedical applications. Their use in biomedical applications such as tissue engineering, photothermal therapy, neural regeneration, and drug delivery has opened new horizons in treatment of age-old ailments. It is also a rapidly developing area of advanced research. A new approach of integrating 3D printing (3DP), a layer-by-layer deposition technique for building structures, along with 2DNM multifunctional inks, has gained considerable attention in recent times, especially in biomedical applications. With the ever-growing demand in healthcare industry for novel, efficient, and rapid technologies for therapeutic treatment methods, 3DP structures of 2DNMs provide vast scope for evolution of a new generation of biomedical devices. Recent advances in 3DP structures of dispersed 2DNM inks with established high-performance biomedical properties are focused on. The advantages of their 3D structures, the sustainable formulation methods of such inks, and their feasible printing methods are also covered. Subsequently, it deals with the therapeutic applications of some already researched 3DP structures of 2DNMs and concludes with highlighting the challenges as well as the future directions of research in this area.
Collapse
Affiliation(s)
- Pritam K Chakraborty
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Kandi, Telangana, 502285, India
- School of Engineering, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Victoria, Hawthorn, 3122, Australia
| | - Jalal Azadmanjiri
- Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technická 5, Prague 6, Prague, 166 28, Czech Republic
| | - Chokkakula L P Pavithra
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Kandi, Telangana, 502285, India
| | - Xiaojian Wang
- Centre for 3D Printing Materials and Additive Manufacturing Technology, Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou, 510632, China
| | - Syed H Masood
- School of Engineering, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Victoria, Hawthorn, 3122, Australia
| | - Suhash Ranjan Dey
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Kandi, Telangana, 502285, India
| | - James Wang
- School of Engineering, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Victoria, Hawthorn, 3122, Australia
| |
Collapse
|
471
|
Li T, Chang J, Zhu Y, Wu C. 3D Printing of Bioinspired Biomaterials for Tissue Regeneration. Adv Healthc Mater 2020; 9:e2000208. [PMID: 32338464 DOI: 10.1002/adhm.202000208] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/14/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
Biological systems, which possess remarkable functions and excellent properties, are gradually becoming a source of inspiration for the fabrication of advanced tissue regeneration biomaterials due to their hierarchical structures and novel compositions. It would be meaningful to learn and transfer the characteristics of creatures to biomaterials design. However, traditional strategies cannot satisfy the design requirements of the complicated bioinspired materials for tissue regeneration. 3D printing, as a rapidly developing new technology that can accurately achieve multimaterial and multiscale fabrication, is capable of optimizing the fabrication of bioinspired materials with complex composition and structure. This review summarizes the recent developments in 3D-printed bioinspired biomaterials for multiple tissue regeneration, and especially highlights the progresses on i) traditional bioinspired designs for biomaterials fabrication, ii) biological composition inspired designs for the 3D-printed biomaterials, and iii) biological structure inspired designs for the 3D-printed biomaterials. Finally, the challenges and prospects for the development of 3D-printed bioinspired biomaterials are discussed.
Collapse
Affiliation(s)
- Tian Li
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, No. 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing, 100049, P. R. China
| | - Jiang Chang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, No. 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing, 100049, P. R. China
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, No. 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing, 100049, P. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, No. 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing, 100049, P. R. China
| |
Collapse
|
472
|
Ning L, Gil CJ, Hwang B, Theus AS, Perez L, Tomov ML, Bauser-Heaton H, Serpooshan V. Biomechanical factors in three-dimensional tissue bioprinting. APPLIED PHYSICS REVIEWS 2020; 7:041319. [PMID: 33425087 PMCID: PMC7780402 DOI: 10.1063/5.0023206] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/23/2020] [Indexed: 05/07/2023]
Abstract
3D bioprinting techniques have shown great promise in various fields of tissue engineering and regenerative medicine. Yet, creating a tissue construct that faithfully represents the tightly regulated composition, microenvironment, and function of native tissues is still challenging. Among various factors, biomechanics of bioprinting processes play fundamental roles in determining the ultimate outcome of manufactured constructs. This review provides a comprehensive and detailed overview on various biomechanical factors involved in tissue bioprinting, including those involved in pre, during, and post printing procedures. In preprinting processes, factors including viscosity, osmotic pressure, and injectability are reviewed and their influence on cell behavior during the bioink preparation is discussed, providing a basic guidance for the selection and optimization of bioinks. In during bioprinting processes, we review the key characteristics that determine the success of tissue manufacturing, including the rheological properties and surface tension of the bioink, printing flow rate control, process-induced mechanical forces, and the in situ cross-linking mechanisms. Advanced bioprinting techniques, including embedded and multi-material printing, are explored. For post printing steps, general techniques and equipment that are used for characterizing the biomechanical properties of printed tissue constructs are reviewed. Furthermore, the biomechanical interactions between printed constructs and various tissue/cell types are elaborated for both in vitro and in vivo applications. The review is concluded with an outlook regarding the significance of biomechanical processes in tissue bioprinting, presenting future directions to address some of the key challenges faced by the bioprinting community.
Collapse
Affiliation(s)
- Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia 30322, USA
| | - Carmen J. Gil
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia 30322, USA
| | - Boeun Hwang
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia 30322, USA
| | - Andrea S. Theus
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia 30322, USA
| | - Lilanni Perez
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia 30322, USA
| | - Martin L. Tomov
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia 30322, USA
| | - Holly Bauser-Heaton
- Authors to whom correspondence should be addressed:. Telephone: 404-712-9717. Fax: 404-727-9873
| | - Vahid Serpooshan
- Authors to whom correspondence should be addressed:. Telephone: 404-712-9717. Fax: 404-727-9873
| |
Collapse
|
473
|
Khoshnood N, Zamanian A. Decellularized extracellular matrix bioinks and their application in skin tissue engineering. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2020.e00095] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
474
|
Daneshmandi L, Shah S, Jafari T, Bhattacharjee M, Momah D, Saveh-Shemshaki N, Lo KWH, Laurencin CT. Emergence of the Stem Cell Secretome in Regenerative Engineering. Trends Biotechnol 2020; 38:1373-1384. [PMID: 32622558 PMCID: PMC7666064 DOI: 10.1016/j.tibtech.2020.04.013] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
The secretome is defined as the set of molecules and biological factors that are secreted by cells into the extracellular space. In the past decade, secretome-based therapies have emerged as a promising approach to overcome the limitations associated with cell-based therapies for tissue and organ regeneration. Considering the growing number of recent publications related to secretome-based therapies, this review takes a step-by-step engineering approach to evaluate the role of the stem cell secretome in regenerative engineering. We discuss the functional benefits of the secretome, the techniques used to engineer the secretome and tailor its therapeutic effects, and the delivery systems and strategies that have been developed to use the secretome for tissue regeneration.
Collapse
Affiliation(s)
- Leila Daneshmandi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Shiv Shah
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA; Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Tahereh Jafari
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Maumita Bhattacharjee
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Deandra Momah
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
| | - Nikoo Saveh-Shemshaki
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Kevin W-H Lo
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Institute of Materials Science, University of Connecticut, Storrs, CT 06269
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA; Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA; Institute of Materials Science, University of Connecticut, Storrs, CT 06269; Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Medicine, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
475
|
Luo Y, Pan H, Jiang J, Zhao C, Zhang J, Chen P, Lin X, Fan S. Desktop-Stereolithography 3D Printing of a Polyporous Extracellular Matrix Bioink for Bone Defect Regeneration. Front Bioeng Biotechnol 2020; 8:589094. [PMID: 33240866 PMCID: PMC7677189 DOI: 10.3389/fbioe.2020.589094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/19/2020] [Indexed: 12/28/2022] Open
Abstract
Introduction Decellularized tendon extracellular matrix (tECM) perfectly provides the natural environment and holds great potential for bone regeneration in Bone tissue engineering (BTE) area. However, its densifying fiber structure leads to reduced cell permeability. Our study aimed to combine tECM with polyethylene glycol diacrylate (PEGDA) to form a biological scaffold with appropriate porosity and strength using stereolithography (SLA) technology for bone defect repair. Methods The tECM was produced and evaluated. Mesenchymal stem cell (MSC) was used to evaluate the biocompatibility of PEGDA/tECM bioink in vitro. Mineralization ability of the bioink was also evaluated in vitro. After preparing 3D printed polyporous PEGDA/tECM scaffolds (3D-pPES) via SLA, the calvarial defect generation capacity of 3D-pPES was assessed. Results The tECM was obtained and the decellularized effect was confirmed. The tECM increased the swelling ratio and porosity of PEGDA bioink, both cellular proliferation and biomineralization in vitro of the bioink were significantly optimized. The 3D-pPES was fabricated. Compared to the control group, increased cell migration efficiency, up-regulation of osteogenic differentiation RNA level, and better calvarial defect repair in rat of the 3D-pPES group were observed. Conclusion This study demonstrates that the 3D-pPES may be a promising strategy for bone defect treatment.
Collapse
Affiliation(s)
- Yunxiang Luo
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hao Pan
- Department of Orthopaedic, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiuzhou Jiang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Chenchen Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jianfeng Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
476
|
Functional 3D printing: Approaches and bioapplications. Biosens Bioelectron 2020; 175:112849. [PMID: 33250333 DOI: 10.1016/j.bios.2020.112849] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/28/2020] [Accepted: 11/22/2020] [Indexed: 12/17/2022]
Abstract
3D printing technology has become a mature manufacturing technique, widely used for its advantages over the traditional methods, such as the end-user customization and rapid prototyping, useful in different application fields, including the biomedical one. Indeed, it represents a helpful tool for the realization of biodevices (i.e. biosensors, microfluidic bioreactors, drug delivery systems and Lab-On-Chip). In this perspective, the development of 3D printable materials with intrinsic functionalities, through the so-called 4D printing, introduces novel opportunities for the fabrication of "smart" or stimuli-responsive devices. Indeed, functional 3D printable materials can modify their surfaces, structures, properties or even shape in response to specific stimuli (such as pressure, temperature or light radiation), adding to the printed object new interesting properties exploited after the fabrication process. In this context, by combining 3D printing technology with an accurate materials' design, functional 3D objects with built-in (bio)chemical functionalities, having biorecognition, biocatalytic and drug delivery capabilities are here reported.
Collapse
|
477
|
Fritschen A, Blaeser A. Biosynthetic, biomimetic, and self-assembled vascularized Organ-on-a-Chip systems. Biomaterials 2020; 268:120556. [PMID: 33310539 DOI: 10.1016/j.biomaterials.2020.120556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
Organ-on-a-Chip (OOC) devices have seen major advances in the last years with respect to biological complexity, physiological composition and biomedical relevance. In this context, integration of vasculature has proven to be a crucial element for long-term culture of thick tissue samples as well as for realistic pharmacokinetic, toxicity and metabolic modelling. With the emergence of digital production technologies and the reinvention of existing tools, a multitude of design approaches for guided angio- and vasculogenesis is available today. The underlying production methods can be categorized into biosynthetic, biomimetic and self-assembled vasculature formation. The diversity and importance of production approaches, vascularization strategies as well as biomaterials and cell sourcing are illustrated in this work. A comprehensive technological review with a strong focus on the challenge of producing physiologically relevant vascular structures is given. Finally, the remaining obstacles and opportunities in the development of vascularized Organ-on-a-Chip platforms for advancing drug development and predictive disease modelling are noted.
Collapse
Affiliation(s)
- Anna Fritschen
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Germany.
| | - Andreas Blaeser
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Germany; Centre for Synthetic Biology, Technical University of Darmstadt, Germany.
| |
Collapse
|
478
|
Schmidt K, Berg J, Roehrs V, Kurreck J, Al-Zeer MA. 3D-bioprinted HepaRG cultures as a model for testing long term aflatoxin B1 toxicity in vitro. Toxicol Rep 2020; 7:1578-1587. [PMID: 33304827 PMCID: PMC7708771 DOI: 10.1016/j.toxrep.2020.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/27/2022] Open
Abstract
In recent years 3D-bioprinting technology has been developed as an alternative to animal testing. It possesses a great potential for in vitro testing as it aims to mimic human organs and physiology. In the present study, an alginate-gelatin-Matrigel based hydrogel was used to prepare 3D-bioprinted HepaRG cultures using a pneumatic extrusion printer. These 3D models were tested for viability and metabolic functions. Using 3D-bioprinted HepaRG cultures, we tested the toxicity of aflatoxin B1 (10 or 20 μM) in vitro and compared the results with 2D HepaRG cultures. There was a dose-dependent toxicity effect on cell viability, reduction of metabolic activity and albumin production. We found that 3D-bioprinted HepaRG cultures are more resistant to aflatoxin B1 treatment than 2D cultures. Although the metabolic activities were reduced upon treatment with aflatoxin B1, the 3D models were still viable and survived longer, up to 3 weeks, than the 2D culture, as visualized by fluorescence microscopy. Furthermore, albumin production recovered slightly in 3D models after one and two weeks of treatment. Taken together, we consider using 3D-bioprinting technology to generate 3D tissue models as an alternative way to study toxicity in vitro and this could also provide a suitable alternative for chronic hepatotoxicity studies in vitro.
Collapse
Affiliation(s)
- Konrad Schmidt
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Johanna Berg
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Viola Roehrs
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Munir A. Al-Zeer
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| |
Collapse
|
479
|
3D Bioprinting of Human Adipose-Derived Stem Cells and Their Tenogenic Differentiation in Clinical-Grade Medium. Int J Mol Sci 2020; 21:ijms21228694. [PMID: 33218011 PMCID: PMC7698777 DOI: 10.3390/ijms21228694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Defining the best combination of cells and biomaterials is a key challenge for the development of tendon tissue engineering (TE) strategies. Adipose-derived stem cells (ASCs) are ideal candidates for this purpose. In addition, controlled cell-based products adherent to good manufacturing practice (GMP) are required for their clinical scale-up. With this aim, in this study, ASC 3D bioprinting and GMP-compliant tenogenic differentiation were investigated. In detail, primary human ASCs were embedded within a nanofibrillar-cellulose/alginate bioink and 3D-bioprinted into multi-layered square-grid matrices. Bioink viscoelastic properties and scaffold ultrastructural morphology were analyzed by rheology and scanning electron microscopy (SEM). The optimal cell concentration for printing among 3, 6 and 9 × 106 ASC/mL was evaluated in terms of cell viability. ASC morphology was characterized by SEM and F-actin immunostaining. Tenogenic differentiation ability was then evaluated in terms of cell viability, morphology and expression of scleraxis and collagen type III by biochemical induction using BMP-12, TGF-β3, CTGF and ascorbic acid supplementation (TENO). Pro-inflammatory cytokine release was also assessed. Bioprinted ASCs showed high viability and survival and exhibited a tenocyte-like phenotype after biochemical induction, with no inflammatory response to the bioink. In conclusion, we report a first proof of concept for the clinical scale-up of ASC 3D bioprinting for tendon TE.
Collapse
|
480
|
3D Printing Decellularized Extracellular Matrix to Design Biomimetic Scaffolds for Skeletal Muscle Tissue Engineering. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2689701. [PMID: 33282941 PMCID: PMC7685790 DOI: 10.1155/2020/2689701] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/08/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
Functional engineered muscles are still a critical clinical issue to be addressed, although different strategies have been considered so far for the treatment of severe muscular injuries. Indeed, the regenerative capacity of skeletal muscle (SM) results inadequate for large-scale defects, and currently, SM reconstruction remains a complex and unsolved task. For this aim, tissue engineered muscles should provide a proper biomimetic extracellular matrix (ECM) alternative, characterized by an aligned/microtopographical structure and a myogenic microenvironment, in order to promote muscle regeneration. As a consequence, both materials and fabrication techniques play a key role to plan an effective therapeutic approach. Tissue-specific decellularized ECM (dECM) seems to be one of the most promising material to support muscle regeneration and repair. 3D printing technologies, on the other side, enable the fabrication of scaffolds with a fine and detailed microarchitecture and patient-specific implants with high structural complexity. To identify innovative biomimetic solutions to develop engineered muscular constructs for the treatment of SM loss, the more recent (last 5 years) reports focused on SM dECM-based scaffolds and 3D printing technologies for SM regeneration are herein reviewed. Possible design inputs for 3D printed SM dECM-based scaffolds for muscular regeneration are also suggested.
Collapse
|
481
|
Jeffries GDM, Xu S, Lobovkina T, Kirejev V, Tusseau F, Gyllensten C, Singh AK, Karila P, Moll L, Orwar O. 3D micro-organisation printing of mammalian cells to generate biological tissues. Sci Rep 2020; 10:19529. [PMID: 33173097 PMCID: PMC7656262 DOI: 10.1038/s41598-020-74191-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/28/2020] [Indexed: 01/06/2023] Open
Abstract
Significant strides have been made in the development of in vitro systems for disease modelling. However, the requirement of microenvironment control has placed limitations on the generation of relevant models. Herein, we present a biological tissue printing approach that employs open-volume microfluidics to position individual cells in complex 2D and 3D patterns, as well as in single cell arrays. The variety of bioprinted cell types employed, including skin epithelial (HaCaT), skin cancer (A431), liver cancer (Hep G2), and fibroblast (3T3-J2) cells, all of which exhibited excellent viability and survivability, allowing printed structures to rapidly develop into confluent tissues. To demonstrate a simple 2D oncology model, A431 and HaCaT cells were printed and grown into tissues. Furthermore, a basic skin model was established to probe drug response. 3D tissue formation was demonstrated by co-printing Hep G2 and 3T3-J2 cells onto an established fibroblast layer, the functionality of which was probed by measuring albumin production, and was found to be higher in comparison to both 2D and monoculture approaches. Bioprinting of primary cells was tested using acutely isolated primary rat dorsal root ganglia neurons, which survived and established processes. The presented technique offers a novel open-volume microfluidics approach to bioprint cells for the generation of biological tissues.
Collapse
Affiliation(s)
| | - Shijun Xu
- Fluicell AB, Flöjelbergsgatan 8C, 431 37, Mölndal, Sweden
| | | | | | | | | | | | - Paul Karila
- Cellectricon AB, Neongatan 4B, 431 53, Mölndal, Sweden
| | - Lydia Moll
- Department of Physiology and Pharmacology, Karolinska Intitutet, Solnavägen 1, 171 77, Solna, Sweden.,Cellectricon AB, Neongatan 4B, 431 53, Mölndal, Sweden
| | - Owe Orwar
- Department of Physiology and Pharmacology, Karolinska Intitutet, Solnavägen 1, 171 77, Solna, Sweden.
| |
Collapse
|
482
|
Vascular bioprinting with enzymatically degradable bioinks via multi-material projection-based stereolithography. Acta Biomater 2020; 117:121-132. [PMID: 32980542 DOI: 10.1016/j.actbio.2020.09.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/02/2020] [Accepted: 09/18/2020] [Indexed: 12/20/2022]
Abstract
Introduction of cavities and channels into 3D bioprinted constructs is a prerequisite for recreating physiological tissue architectures and integrating vasculature. Projection-based stereolithography inherently offers high printing speed with high spatial resolution, but so far has been incapable of fabricating complex native tissue architectures with cellular and biomaterial diversity. The use of sacrificial photoinks, i.e. photopolymerisable biomaterials that can be removed after printing, theoretically allows for the creation of any construct geometry via a multi-material printing process. However, the realisation of this strategy has been challenging because of difficult technical implementation and a lack of performant biomaterials. In this work, we use our projection-based, multi-material stereolithographic bioprinter and an enzymatically degradable sacrificial photoink to overcome the current hurdles. Multiple, hyaluronic acid-based photoinks were screened for printability, mechanical properties and digestibility through hyaluronidase. A formulation meeting all major requirements, i.e. desirable printing properties, generation of sufficiently strong hydrogels, as well as fast digestion rate, was identified. Biocompatibility of the material system was confirmed by embedding of human umbilical vein endothelial cells with followed enzymatic release. As a proof-of-concept, we bioprinted vascular models containing perfusable, endothelial cell-lined channels that remained stable for 28 days in culture. Our work establishes digestible sacrificial biomaterials as a new material strategy for 3D bioprinting of complex tissue models.
Collapse
|
483
|
Chen K, Chen X, Han X, Fu Y. A comparison study on the release kinetics and mechanism of bovine serum albumin and nanoencapsulated albumin from hydrogel networks. Int J Biol Macromol 2020; 163:1291-1300. [DOI: 10.1016/j.ijbiomac.2020.07.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022]
|
484
|
Shah Mohammadi M, Buchen JT, Pasquina PF, Niklason LE, Alvarez LM, Jariwala SH. Critical Considerations for Regeneration of Vascularized Composite Tissues. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:366-381. [PMID: 33115331 DOI: 10.1089/ten.teb.2020.0223] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Effective vascularization is vital for survival and functionality of complex tissue-engineered organs. The formation of the microvasculature, composed of endothelial cells (ECs) alone, has been mostly used to restore the vascular networks in organs. However, recent heterocellular studies demonstrate that co-culturing is a more effective approach in revascularization of engineered organs. This review presents key considerations for manufacturing of artificial vascularized composite tissues. We summarize the importance of co-cultures and the multicellular interactions with ECs, as well as design and use of bioreactors, as critical considerations for tissue vascularization. In addition, as an emerging scaffolding technique, this review also highlights the current caveats and hurdles associated with three-dimensional bioprinting and discusses recent developments in bioprinting strategies such as four-dimensional bioprinting and its future outlook for manufacturing of vascularized tissue constructs. Finally, the review concludes with addressing the critical challenges in the regulatory pathway and clinical translation of artificial composite tissue grafts. Impact statement Regeneration of composite tissues is critical as biophysical and biochemical characteristics differ between various types of tissues. Engineering a vascularized composite tissue has remained unresolved and requires additional evaluations along with optimization of methodologies and standard operating procedures. To this end, the main hurdle is creating a viable vascular endothelium that remains functional for a longer duration postimplantation, and can be manufactured using clinically appropriate source of cell lines that are scalable in vitro for the fabrication of human-scale organs. This review presents key considerations for regeneration and manufacturing of vascularized composite tissues as the field advances.
Collapse
Affiliation(s)
- Maziar Shah Mohammadi
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Jack T Buchen
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Paul F Pasquina
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Laura E Niklason
- Department of Anesthesia and Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Luis M Alvarez
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Lung Biotechnology PBC, Silver Spring, Maryland, USA
| | - Shailly H Jariwala
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
485
|
3D Printing and NIR Fluorescence Imaging Techniques for the Fabrication of Implants. MATERIALS 2020; 13:ma13214819. [PMID: 33126650 PMCID: PMC7662749 DOI: 10.3390/ma13214819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/19/2020] [Accepted: 10/27/2020] [Indexed: 12/17/2022]
Abstract
Three-dimensional (3D) printing technology holds great potential to fabricate complex constructs in the field of regenerative medicine. Researchers in the surgical fields have used 3D printing techniques and their associated biomaterials for education, training, consultation, organ transplantation, plastic surgery, surgical planning, dentures, and more. In addition, the universal utilization of 3D printing techniques enables researchers to exploit different types of hardware and software in, for example, the surgical fields. To realize the 3D-printed structures to implant them in the body and tissue regeneration, it is important to understand 3D printing technology and its enabling technologies. This paper concisely reviews 3D printing techniques in terms of hardware, software, and materials with a focus on surgery. In addition, it reviews bioprinting technology and a non-invasive monitoring method using near-infrared (NIR) fluorescence, with special attention to the 3D-bioprinted tissue constructs. NIR fluorescence imaging applied to 3D printing technology can play a significant role in monitoring the therapeutic efficacy of 3D structures for clinical implants. Consequently, these techniques can provide individually customized products and improve the treatment outcome of surgeries.
Collapse
|
486
|
Farhat W, Chatelain F, Marret A, Faivre L, Arakelian L, Cattan P, Fuchs A. Trends in 3D bioprinting for esophageal tissue repair and reconstruction. Biomaterials 2020; 267:120465. [PMID: 33129189 DOI: 10.1016/j.biomaterials.2020.120465] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 02/08/2023]
Abstract
In esophageal pathologies, such as esophageal atresia, cancers, caustic burns, or post-operative stenosis, esophageal replacement is performed by using parts of the gastrointestinal tract to restore nutritional autonomy. However, this surgical procedure most often does not lead to complete functional recovery and is instead associated with many complications resulting in a decrease in the quality of life and survival rate. Esophageal tissue engineering (ETE) aims at repairing the defective esophagus and is considered as a promising therapeutic alternative. Noteworthy progress has recently been made in the ETE research area but strong challenges remain to replicate the structural and functional integrity of the esophagus with the approaches currently being developed. Within this context, 3D bioprinting is emerging as a new technology to facilitate the patterning of both cellular and acellular bioinks into well-organized 3D functional structures. Here, we present a comprehensive overview of the recent advances in tissue engineering for esophageal reconstruction with a specific focus on 3D bioprinting approaches in ETE. Current biofabrication techniques and bioink features are highlighted, and these are discussed in view of the complexity of the native esophagus that the designed substitute needs to replace. Finally, perspectives on recent strategies for fabricating other tubular organ substitutes via 3D bioprinting are discussed briefly for their potential in ETE applications.
Collapse
Affiliation(s)
- Wissam Farhat
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France
| | - François Chatelain
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France
| | - Auriane Marret
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France
| | - Lionel Faivre
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; Assistance Publique - Hôpitaux de Paris, Unité de Thérapie Cellulaire, Hôpital Saint-Louis, Paris, France
| | - Lousineh Arakelian
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; Assistance Publique - Hôpitaux de Paris, Unité de Thérapie Cellulaire, Hôpital Saint-Louis, Paris, France
| | - Pierre Cattan
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; Assistance Publique - Hôpitaux de Paris, Service de Chirurgie Digestive, Hôpital Saint-Louis, Paris, France
| | - Alexandra Fuchs
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France.
| |
Collapse
|
487
|
Ruiz-Garcia H, Alvarado-Estrada K, Schiapparelli P, Quinones-Hinojosa A, Trifiletti DM. Engineering Three-Dimensional Tumor Models to Study Glioma Cancer Stem Cells and Tumor Microenvironment. Front Cell Neurosci 2020; 14:558381. [PMID: 33177991 PMCID: PMC7596188 DOI: 10.3389/fncel.2020.558381] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is the most common and devastating primary brain tumor, leading to a uniform fatality after diagnosis. A major difficulty in eradicating GBM is the presence of microscopic residual infiltrating disease remaining after multimodality treatment. Glioma cancer stem cells (CSCs) have been pinpointed as the treatment-resistant tumor component that seeds ultimate tumor progression. Despite the key role of CSCs, the ideal preclinical model to study the genetic and epigenetic landmarks driving their malignant behavior while simulating an accurate interaction with the tumor microenvironment (TME) is still missing. The introduction of three-dimensional (3D) tumor platforms, such as organoids and 3D bioprinting, has allowed for a better representation of the pathophysiologic interactions between glioma CSCs and the TME. Thus, these technologies have enabled a more detailed study of glioma biology, tumor angiogenesis, treatment resistance, and even performing high-throughput screening assays of drug susceptibility. First, we will review the foundation of glioma biology and biomechanics of the TME, and then the most up-to-date insights about the applicability of these new tools in malignant glioma research.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States.,Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Paula Schiapparelli
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Daniel M Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States.,Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
488
|
Abstract
The field of tissue engineering has advanced over the past decade, but the largest impact on human health should be achieved with the transition of engineered solid organs to the clinic. The number of patients suffering from solid organ disease continues to increase, with over 100 000 patients on the U.S. national waitlist and approximately 730 000 deaths in the United States resulting from end-stage organ disease annually. While flat, tubular, and hollow nontubular engineered organs have already been implanted in patients, in vitro formation of a fully functional solid organ at a translatable scale has not yet been achieved. Thus, one major goal is to bioengineer complex, solid organs for transplantation, composed of patient-specific cells. Among the myriad of approaches attempted to engineer solid organs, 3D bioprinting offers unmatched potential. This review highlights the structural complexity which must be engineered at nano-, micro-, and mesostructural scales to enable organ function. We showcase key advances in bioprinting solid organs with complex vascular networks and functioning microstructures, advances in biomaterials science that have enabled this progress, the regulatory hurdles the field has yet to overcome, and cutting edge technologies that bring us closer to the promise of engineered solid organs.
Collapse
Affiliation(s)
- Adam M Jorgensen
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
489
|
Theus AS, Ning L, Hwang B, Gil C, Chen S, Wombwell A, Mehta R, Serpooshan V. Bioprintability: Physiomechanical and Biological Requirements of Materials for 3D Bioprinting Processes. Polymers (Basel) 2020; 12:E2262. [PMID: 33019639 PMCID: PMC7599870 DOI: 10.3390/polym12102262] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022] Open
Abstract
Three-dimensional (3D) bioprinting is an additive manufacturing process that utilizes various biomaterials that either contain or interact with living cells and biological systems with the goal of fabricating functional tissue or organ mimics, which will be referred to as bioinks. These bioinks are typically hydrogel-based hybrid systems with many specific features and requirements. The characterizing and fine tuning of bioink properties before, during, and after printing are therefore essential in developing reproducible and stable bioprinted constructs. To date, myriad computational methods, mechanical testing, and rheological evaluations have been used to predict, measure, and optimize bioinks properties and their printability, but none are properly standardized. There is a lack of robust universal guidelines in the field for the evaluation and quantification of bioprintability. In this review, we introduced the concept of bioprintability and discussed the significant roles of various physiomechanical and biological processes in bioprinting fidelity. Furthermore, different quantitative and qualitative methodologies used to assess bioprintability will be reviewed, with a focus on the processes related to pre, during, and post printing. Establishing fully characterized, functional bioink solutions would be a big step towards the effective clinical applications of bioprinted products.
Collapse
Affiliation(s)
- Andrea S. Theus
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Boeun Hwang
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Carmen Gil
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Shuai Chen
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Allison Wombwell
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Riya Mehta
- Department of Biology, Emory University, Atlanta, GA 30322, USA;
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
490
|
Farzan A, Borandeh S, Zanjanizadeh Ezazi N, Lipponen S, Santos HA, Seppälä J. 3D scaffolding of fast photocurable polyurethane for soft tissue engineering by stereolithography: Influence of materials and geometry on growth of fibroblast cells. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.109988] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
491
|
Fu L, Yang Z, Gao C, Li H, Yuan Z, Wang F, Sui X, Liu S, Guo Q. Advances and prospects in biomimetic multilayered scaffolds for articular cartilage regeneration. Regen Biomater 2020; 7:527-542. [PMID: 33365139 PMCID: PMC7748444 DOI: 10.1093/rb/rbaa042] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/13/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Due to the sophisticated hierarchical structure and limited reparability of articular cartilage (AC), the ideal regeneration of AC defects has been a major challenge in the field of regenerative medicine. As defects progress, they often extend from the cartilage layer to the subchondral bone and ultimately lead to osteoarthritis. Tissue engineering techniques bring new hope for AC regeneration. To meet the regenerative requirements of the heterogeneous and layered structure of native AC tissue, a substantial number of multilayered biomimetic scaffolds have been studied. Ideal multilayered scaffolds should generate zone-specific functional tissue similar to native AC tissue. This review focuses on the current status of multilayered scaffolds developed for AC defect repair, including design strategies based on the degree of defect severity and the zone-specific characteristics of AC tissue, the selection and composition of biomaterials, and techniques for design and manufacturing. The challenges and future perspectives of biomimetic multilayered scaffold strategies for AC regeneration are also discussed.
Collapse
Affiliation(s)
- Liwei Fu
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, China.,Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Zhen Yang
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, China.,Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Cangjian Gao
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, China.,Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Hao Li
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, China.,Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Zhiguo Yuan
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China.,Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No. 160 Pujian Road, Pudong New District, Shanghai 200127, China
| | - Fuxin Wang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xiang Sui
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shuyun Liu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Quanyi Guo
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, China.,Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| |
Collapse
|
492
|
Zhang Y, Leng H, Du Z, Huang Y, Liu X, Zhao Z, Zhang X, Cai Q, Yang X. Efficient regeneration of rat calvarial defect with gelatin-hydroxyapatite composite cryogel. Biomed Mater 2020; 15:065005. [DOI: 10.1088/1748-605x/ab9422] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
493
|
Wang Z, Hui A, Zhao H, Ye X, Zhang C, Wang A, Zhang C. A Novel 3D-bioprinted Porous Nano Attapulgite Scaffolds with Good Performance for Bone Regeneration. Int J Nanomedicine 2020; 15:6945-6960. [PMID: 33061361 PMCID: PMC7520466 DOI: 10.2147/ijn.s254094] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 08/05/2020] [Indexed: 12/25/2022] Open
Abstract
Background Natural clay nanomaterials are an emerging class of biomaterial with great potential for tissue engineering and regenerative medicine applications, most notably for osteogenesis. Materials and Methods Herein, for the first time, novel tissue engineering scaffolds were prepared by 3D bioprinter using nontoxic and bioactive natural attapulgite (ATP) nanorods as starting materials, with polyvinyl alcohol as binder, and then sintered to obtain final scaffolds. The microscopic morphology and structure of ATP particles and scaffolds were observed by transmission electron microscope and scanning electron microscope. In vitro biocompatibility and osteogenesis with osteogenic precursor cell (hBMSCs) were assayed using MTT method, Live/Dead cell staining, alizarin red staining and RT-PCR. In vivo bone regeneration was evaluated with micro-CT and histology analysis in rat cranium defect model. Results We successfully printed a novel porous nano-ATP scaffold designed with inner channels with a dimension of 500 µm and wall structures with a thickness of 330 µm. The porosity of current 3D-printed scaffolds ranges from 75% to 82% and the longitudinal compressive strength was up to 4.32±0.52 MPa. We found firstly that nano-ATP scaffolds with excellent biocompatibility for hBMSCscould upregulate the expression of osteogenesis-related genes bmp2 and runx2 and calcium deposits in vitro. Interestingly, micro-CT and histology analysis revealed abundant newly formed bone was observed along the defect margin, even above and within the 3D bioprinted porous ATP scaffolds in a rat cranial defect model. Furthermore, histology analysis demonstrated that bone was formed directly following a process similar to membranous ossification without any intermediate cartilage formation and that many newly formed blood vessels are within the pores of 3D-printed scaffolds at four and eight weeks. Conclusion These results suggest that the 3D-printed porous nano-ATP scaffolds are promising candidates for bone tissue engineering by osteogenesis and angiogenesis.
Collapse
Affiliation(s)
- Zehao Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| | - Aiping Hui
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-Material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China
| | - Hongbin Zhao
- Changzhou No. 2 People's Hospital, Changzhou 213000, People's Republic of China
| | - Xiaohan Ye
- Beijing Tiantan Biological Products Co., Ltd, Beijing 100000, People's Republic of China
| | - Chao Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| | - Aiqin Wang
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-Material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China
| | - Changqing Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| |
Collapse
|
494
|
Ghorbani F, Li D, Zhong Z, Sahranavard M, Qian Z, Ni S, Zhang Z, Zamanian A, Yu B. Bioprinting a cell‐laden matrix for bone regeneration: A focused review. J Appl Polym Sci 2020. [DOI: 10.1002/app.49888] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Farnaz Ghorbani
- Department of Orthopedics, Shanghai Pudong Hospital Fudan University Pudong Medical Center Shanghai China
| | - Dejian Li
- Department of Orthopedics, Shanghai Pudong Hospital Fudan University Pudong Medical Center Shanghai China
| | - Zeyuan Zhong
- Department of Orthopedics, Shanghai Pudong Hospital Fudan University Pudong Medical Center Shanghai China
| | - Melika Sahranavard
- Department of Nanotechnology and Advanced Materials Materials and Energy Research Center Karaj Iran
| | - Zhi Qian
- Department of Orthopedics, Shanghai Pudong Hospital Fudan University Pudong Medical Center Shanghai China
| | - Shuo Ni
- Department of Orthopedics, Shanghai Pudong Hospital Fudan University Pudong Medical Center Shanghai China
| | - Zhenhua Zhang
- Department of Orthopedics, Shanghai Pudong Hospital Fudan University Pudong Medical Center Shanghai China
- School of Materials Science and Engineering University of Shanghai for Science and Technology Shanghai China
| | - Ali Zamanian
- Department of Nanotechnology and Advanced Materials Materials and Energy Research Center Karaj Iran
| | - Baoqing Yu
- Department of Orthopedics, Shanghai Pudong Hospital Fudan University Pudong Medical Center Shanghai China
| |
Collapse
|
495
|
Bae M, Yi HG, Jang J, Cho DW. Microphysiological Systems for Neurodegenerative Diseases in Central Nervous System. MICROMACHINES 2020; 11:E855. [PMID: 32947879 PMCID: PMC7570039 DOI: 10.3390/mi11090855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Abstract
Neurodegenerative diseases are among the most severe problems in aging societies. Various conventional experimental models, including 2D and animal models, have been used to investigate the pathogenesis of (and therapeutic mechanisms for) neurodegenerative diseases. However, the physiological gap between humans and the current models remains a hurdle to determining the complexity of an irreversible dysfunction in a neurodegenerative disease. Therefore, preclinical research requires advanced experimental models, i.e., those more physiologically relevant to the native nervous system, to bridge the gap between preclinical stages and patients. The neural microphysiological system (neural MPS) has emerged as an approach to summarizing the anatomical, biochemical, and pathological physiology of the nervous system for investigation of neurodegenerative diseases. This review introduces the components (such as cells and materials) and fabrication methods for designing a neural MPS. Moreover, the review discusses future perspectives for improving the physiological relevance to native neural systems.
Collapse
Affiliation(s)
- Mihyeon Bae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
| | - Hee-Gyeong Yi
- Department of Rural and Biosystems Engineering, College of Agricultural Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| |
Collapse
|
496
|
Next-generation tissue-engineered heart valves with repair, remodelling and regeneration capacity. Nat Rev Cardiol 2020; 18:92-116. [PMID: 32908285 DOI: 10.1038/s41569-020-0422-8] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2020] [Indexed: 02/06/2023]
Abstract
Valvular heart disease is a major cause of morbidity and mortality worldwide. Surgical valve repair or replacement has been the standard of care for patients with valvular heart disease for many decades, but transcatheter heart valve therapy has revolutionized the field in the past 15 years. However, despite the tremendous technical evolution of transcatheter heart valves, to date, the clinically available heart valve prostheses for surgical and transcatheter replacement have considerable limitations. The design of next-generation tissue-engineered heart valves (TEHVs) with repair, remodelling and regenerative capacity can address these limitations, and TEHVs could become a promising therapeutic alternative for patients with valvular disease. In this Review, we present a comprehensive overview of current clinically adopted heart valve replacement options, with a focus on transcatheter prostheses. We discuss the various concepts of heart valve tissue engineering underlying the design of next-generation TEHVs, focusing on off-the-shelf technologies. We also summarize the latest preclinical and clinical evidence for the use of these TEHVs and describe the current scientific, regulatory and clinical challenges associated with the safe and broad clinical translation of this technology.
Collapse
|
497
|
Pahlevanzadeh F, Mokhtari H, Bakhsheshi-Rad HR, Emadi R, Kharaziha M, Valiani A, Poursamar SA, Ismail AF, RamaKrishna S, Berto F. Recent Trends in Three-Dimensional Bioinks Based on Alginate for Biomedical Applications. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E3980. [PMID: 32911867 PMCID: PMC7557490 DOI: 10.3390/ma13183980] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023]
Abstract
Three-dimensional (3D) bioprinting is an appealing and revolutionary manufacturing approach for the accurate placement of biologics, such as living cells and extracellular matrix (ECM) components, in the form of a 3D hierarchical structure to fabricate synthetic multicellular tissues. Many synthetic and natural polymers are applied as cell printing bioinks. One of them, alginate (Alg), is an inexpensive biomaterial that is among the most examined hydrogel materials intended for vascular, cartilage, and bone tissue printing. It has also been studied pertaining to the liver, kidney, and skin, due to its excellent cell response and flexible gelation preparation through divalent ions including calcium. Nevertheless, Alg hydrogels possess certain negative aspects, including weak mechanical characteristics, poor printability, poor structural stability, and poor cell attachment, which may restrict its usage along with the 3D printing approach to prepare artificial tissue. In this review paper, we prepare the accessible materials to be able to encourage and boost new Alg-based bioink formulations with superior characteristics for upcoming purposes in drug delivery systems. Moreover, the major outcomes are discussed, and the outstanding concerns regarding this area and the scope for upcoming examination are outlined.
Collapse
Affiliation(s)
- Farnoosh Pahlevanzadeh
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
- Department of Anatomical Science, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Hamidreza Mokhtari
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Hamid Reza Bakhsheshi-Rad
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Rahmatollah Emadi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Ali Valiani
- Department of Anatomical Science, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - S Ali Poursamar
- Biomaterials, Nanotechnology, and Tissue Engineering Group, Advanced Medical Technology Department, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Ahmad Fauzi Ismail
- Advanced Membrane Technology Research Center (AMTEC), Universiti Teknologi Malaysia, Skudai 81310, Johor Bahru, Johor, Malaysia
| | - Seeram RamaKrishna
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Filippo Berto
- Department of Mechanical and Industrial Engineering, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
498
|
Toledo Stuani VD, do Prado Manfredi GG, Miyahara Kondo VA, Noritomi PY, Lisboa-Filho PN, Passanezi Sant’Ana AC. The use of additively manufactured scaffolds for treating gingival recession associated with interproximal defects. ACTA ACUST UNITED AC 2020. [DOI: 10.2217/3dp-2020-0008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gingival recessions are a highly prevalent issue that is often associated with interproximal tissue deficiency. An intervention in these scenarios is of extreme importance since these defects can lead to aesthetic, phonetic and other dental problems. Unfortunately, the treatment of advanced gingival recessions is a major challenge in periodontics because of its unpredictability. In such cases, the use of injectable fillings, connective tissue grafts or bone grafts for vertical regeneration in interproximal area presents limited results. Considering that, this special report reviewed the possible use of additively manufactured scaffolds as a therapeutic option. A 3D-printed personalized therapy is expected to simplify the regeneration of interproximal area, enabling bone regeneration, new papilla formation and root coverage.
Collapse
Affiliation(s)
- Vitor de Toledo Stuani
- Discipline of Periodontology, Bauru School of Dentistry – University of Sao Paulo, Bauru, Brazil
| | | | | | - Pedro Yoshito Noritomi
- Nucleus of Three-Dimensional Technologies (NT3D), Center for Information Technology Renato Archer, Campinas, Brazil
| | | | | |
Collapse
|
499
|
Liu H, Zheng G, Cheng X, Yang X, Zhao G. Simulation Analysis of the Influence of Nozzle Structure Parameters on Material Controllability. MICROMACHINES 2020; 11:E826. [PMID: 32878235 PMCID: PMC7570424 DOI: 10.3390/mi11090826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/24/2020] [Accepted: 08/29/2020] [Indexed: 11/16/2022]
Abstract
With the evolution of three-dimensional (3D) printing, many restrictive factors of 3D printing have been explored to upgrade the feasibility of 3D printing technology, such as nozzle structure, print resolution, cell viability, etc., which has attracted extensive attention due to its possibility of curing disease in tissue engineering and organ regeneration. In this paper, we have developed a novel nozzle for 3D printing, numerical simulation, and finite element analysis have been used to optimize the nozzle structure and further clarified the influence of nozzle structure parameters on material controllability. Using novel nozzle structure, we firstly adopt ANSYS-FLUENT to analyze material controllability under the different inner cavity diameter, outer cavity diameter and lead length. Secondly, the orthogonal experiments with the novel nozzle are carried out in order to verify the influence law of inner cavity diameter, outer cavity diameter, and lead length under all sorts of conditions. The experiment results show that the material P diameter can be controlled by changing the parameters. The influence degree of parameters on material P diameter is shown that lead length > inner cavity diameter > outer cavity diameter. Finally, the optimized parameters of nozzle structure have been adjusted to estimate the material P diameter in 3D printing.
Collapse
Affiliation(s)
- Huanbao Liu
- School of Mechanical Engineering, Shandong University of Technology, Zibo 255000, China; (G.Z.); (X.C.); (G.Z.)
| | - Guangming Zheng
- School of Mechanical Engineering, Shandong University of Technology, Zibo 255000, China; (G.Z.); (X.C.); (G.Z.)
| | - Xiang Cheng
- School of Mechanical Engineering, Shandong University of Technology, Zibo 255000, China; (G.Z.); (X.C.); (G.Z.)
| | - Xianhai Yang
- Analytical Testing Center, Shandong University of Technology, Zibo 255000, China;
| | - Guangxi Zhao
- School of Mechanical Engineering, Shandong University of Technology, Zibo 255000, China; (G.Z.); (X.C.); (G.Z.)
| |
Collapse
|
500
|
Shpichka A, Bikmulina P, Peshkova M, Kosheleva N, Zurina I, Zahmatkesh E, Khoshdel-Rad N, Lipina M, Golubeva E, Butnaru D, Svistunov A, Vosough M, Timashev P. Engineering a Model to Study Viral Infections: Bioprinting, Microfluidics, and Organoids to Defeat Coronavirus Disease 2019 (COVID-19). Int J Bioprint 2020; 6:302. [PMID: 33089000 PMCID: PMC7557357 DOI: 10.18063/ijb.v6i4.302] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023] Open
Abstract
While the number of studies related to severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) is constantly growing, it is essential to provide a framework of modeling viral infections. Therefore, this review aims to describe the background presented by earlier used models for viral studies and an approach to design an "ideal" tissue model for SARS-CoV-2 infection. Due to the previous successful achievements in antiviral research and tissue engineering, combining the emerging techniques such as bioprinting, microfluidics, and organoid formation are considered to be one of the best approaches to form in vitro tissue models. The fabrication of an integrated multi-tissue bioprinted platform tailored for SARS-CoV-2 infection can be a great breakthrough that can help defeat coronavirus disease in 2019.
Collapse
Affiliation(s)
- Anastasia Shpichka
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Polina Bikmulina
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Maria Peshkova
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Nastasia Kosheleva
- Department of Molecular and Cell Pathophysiology, FSBSI Institute of General Pathology and Pathophysiology, Moscow, Russia
- Department of Embryology, Lomonosov Moscow State University, Faculty of Biology, Moscow, Russia
| | - Irina Zurina
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Department of Molecular and Cell Pathophysiology, FSBSI Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Ensieh Zahmatkesh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel-Rad
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marina Lipina
- Department of Traumatology, Orthopedics and Disaster Surgery, Sechenov University, Moscow, Russia
| | - Elena Golubeva
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Denis Butnaru
- Rector’s Office, Sechenov University, Moscow, Russia
| | | | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
- Department of Polymers and Composites, NN Semenov Institute of Chemical Physics, Moscow, Russia
| |
Collapse
|