101
|
Srivastava N, Choudhury AR. Stimuli-Responsive Polysaccharide-Based Smart Hydrogels and Their Emerging Applications. Ind Eng Chem Res 2022. [DOI: 10.1021/acs.iecr.2c02779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Nandita Srivastava
- Biochemical Engineering Research & Process Development Centre (BERPDC), Institute of Microbial Technology (IMTECH), Council of Scientific and Industrial Research (CSIR), Sector 39A, Chandigarh 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anirban Roy Choudhury
- Biochemical Engineering Research & Process Development Centre (BERPDC), Institute of Microbial Technology (IMTECH), Council of Scientific and Industrial Research (CSIR), Sector 39A, Chandigarh 160036, India
| |
Collapse
|
102
|
Hydrogels and biohydrogels: investigation of origin of production, production methods, and application. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04580-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
103
|
Taymour R, Chicaiza-Cabezas NA, Gelinsky M, Lode A. Core-shell bioprinting of vascularized in vitro liver sinusoid models. Biofabrication 2022; 14. [PMID: 36070706 DOI: 10.1088/1758-5090/ac9019] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/07/2022] [Indexed: 11/12/2022]
Abstract
In vitro liver models allow the investigation of the cell behavior in disease conditions or in response to changes in the microenvironment. A major challenge in liver tissue engineering is to mimic the tissue-level complexity: Besides the selection of suitable biomaterial(s) replacing the extracellular matrix (ECM) and cell sources, the three-dimensional (3D) microarchitecture defined by the fabrication method is a critical factor to achieve functional constructs. In this study, coaxial extrusion-based 3D bioprinting has been applied to develop a liver sinusoid-like model that consists of a core compartment containing pre-vascular structures and a shell compartment containing hepatocytes. The shell ink was composed of alginate and methylcellulose (algMC), dissolved in human fresh frozen plasma. The algMC blend conferred high printing fidelity and stability to the core-shell constructs and the plasma as biologically active component enhanced viability and supported cluster formation and biomarker expression of HepG2 embedded in the shell. For the core, a natural ECM-like ink based on angiogenesis-supporting collagen-fibrin (CF) matrices was developed; the addition of gelatin (G) enabled 3D printing in combination with the plasma-algMC shell ink. Human endothelial cells (HUVEC), laden in the CFG core ink together with human fibroblasts as supportive cells, formed a pre-vascular network in the core in the absence and presence of HepG2 in the shell. The cellular interactions occurring in the triple culture model enhanced the albumin secretion. In conclusion, core-shell bioprinting was shown to be a valuable tool to study cell-cell-interactions and to develop complex tissue-like models.
Collapse
Affiliation(s)
- Rania Taymour
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine, Dresden University of Technology, Fetscherstrasse 74, Dresden, Sachsen, 01307, GERMANY
| | - Nathaly Alejandra Chicaiza-Cabezas
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitaet Dresden, Fetscherstrasse 74, Dresden, Sachsen, 01307, GERMANY
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitat Dresden, Fetscherstr. 74, Dresden, 01062, GERMANY
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitaet Dresden, Fetscherstrasse 74, Dresden, 01307, GERMANY
| |
Collapse
|
104
|
Rizzo R, Bonato A, Chansoria P, Zenobi-Wong M. Macroporous Aligned Hydrogel Microstrands for 3D Cell Guidance. ACS Biomater Sci Eng 2022; 8:3871-3882. [PMID: 35977074 DOI: 10.1021/acsbiomaterials.2c00370] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tissue engineering strongly relies on the use of hydrogels as highly hydrated 3D matrices to support the maturation of laden cells. However, because of the lack of microarchitecture and sufficient porosity, common hydrogel systems do not provide physical cell-instructive guidance cues and efficient transport of nutrients and oxygen to the inner part of the construct. A controlled, organized cellular alignment and resulting alignment of secreted ECM are hallmarks of muscle, tendons, and nerves and play an important role in determining their functional properties. Although several strategies to induce cellular alignment have been investigated in 2D systems, the generation of cell-instructive 3D hydrogels remains a challenge. Here, we report on the development of a simple and scalable method to efficiently generate highly macroporous constructs featuring aligned guidance cues. A precross-linked bulk hydrogel is pressed through a grid with variable opening sizes, thus deconstructing it into an array of aligned, high aspect ratio microgels (microstrands) with tunable diameter that are eventually stabilized by a second photoclick cross-linking step. This method has been investigated and optimized both in silico and in vitro, thereby leading to conditions with excellent viability and organized cellular alignment. Finally, as proof of concept, the method has been shown to direct aligned muscle tissue maturation. These findings demonstrate the 3D physical guidance potential of our system, which can be used for a variety of anisotropic tissues and applications.
Collapse
Affiliation(s)
- Riccardo Rizzo
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich 8093, Switzerland
| | - Angela Bonato
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich 8093, Switzerland
| | - Parth Chansoria
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich 8093, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich 8093, Switzerland
| |
Collapse
|
105
|
Sánchez-Cid P, Jiménez-Rosado M, Romero A, Pérez-Puyana V. Novel Trends in Hydrogel Development for Biomedical Applications: A Review. Polymers (Basel) 2022; 14:polym14153023. [PMID: 35893984 PMCID: PMC9370620 DOI: 10.3390/polym14153023] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 12/11/2022] Open
Abstract
Nowadays, there are still numerous challenges for well-known biomedical applications, such as tissue engineering (TE), wound healing and controlled drug delivery, which must be faced and solved. Hydrogels have been proposed as excellent candidates for these applications, as they have promising properties for the mentioned applications, including biocompatibility, biodegradability, great absorption capacity and tunable mechanical properties. However, depending on the material or the manufacturing method, the resulting hydrogel may not be up to the specific task for which it is designed, thus there are different approaches proposed to enhance hydrogel performance for the requirements of the application in question. The main purpose of this review article was to summarize the most recent trends of hydrogel technology, going through the most used polymeric materials and the most popular hydrogel synthesis methods in recent years, including different strategies of enhancing hydrogels’ properties, such as cross-linking and the manufacture of composite hydrogels. In addition, the secondary objective of this review was to briefly discuss other novel applications of hydrogels that have been proposed in the past few years which have drawn a lot of attention.
Collapse
Affiliation(s)
| | | | - Alberto Romero
- Correspondence: (P.S.-C.); (A.R.); Tel.: +34-954557179 (A.R.)
| | | |
Collapse
|
106
|
Ashammakhi N, GhavamiNejad A, Tutar R, Fricker A, Roy I, Chatzistavrou X, Hoque Apu E, Nguyen KL, Ahsan T, Pountos I, Caterson EJ. Highlights on Advancing Frontiers in Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:633-664. [PMID: 34210148 PMCID: PMC9242713 DOI: 10.1089/ten.teb.2021.0012] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/15/2021] [Indexed: 01/05/2023]
Abstract
The field of tissue engineering continues to advance, sometimes in exponential leaps forward, but also sometimes at a rate that does not fulfill the promise that the field imagined a few decades ago. This review is in part a catalog of success in an effort to inform the process of innovation. Tissue engineering has recruited new technologies and developed new methods for engineering tissue constructs that can be used to mitigate or model disease states for study. Key to this antecedent statement is that the scientific effort must be anchored in the needs of a disease state and be working toward a functional product in regenerative medicine. It is this focus on the wildly important ideas coupled with partnered research efforts within both academia and industry that have shown most translational potential. The field continues to thrive and among the most important recent developments are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies that warrant special attention. Developments in the aforementioned areas as well as future directions are highlighted in this article. Although several early efforts have not come to fruition, there are good examples of commercial profitability that merit continued investment in tissue engineering. Impact statement Tissue engineering led to the development of new methods for regenerative medicine and disease models. Among the most important recent developments in tissue engineering are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies. These technologies and an understanding of them will have impact on the success of tissue engineering and its translation to regenerative medicine. Continued investment in tissue engineering will yield products and therapeutics, with both commercial importance and simultaneous disease mitigation.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, Michigan, USA
| | - Amin GhavamiNejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Annabelle Fricker
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Ipsita Roy
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Xanthippi Chatzistavrou
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Ehsanul Hoque Apu
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
| | - Kim-Lien Nguyen
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Taby Ahsan
- RoosterBio, Inc., Frederick, Maryland, USA
| | - Ippokratis Pountos
- Academic Department of Trauma and Orthopaedics, University of Leeds, Leeds, United Kingdom
| | - Edward J. Caterson
- Division of Plastic Surgery, Department of Surgery, Nemours/Alfred I. du Pont Hospital for Children, Wilmington, Delaware, USA
| |
Collapse
|
107
|
Ji S, Li X, Wang S, Li H, Duan H, Yang X, Lv P. Physically Entangled Anti-Swelling Hydrogels with High Stiffness. Macromol Rapid Commun 2022; 43:e2200272. [PMID: 35640021 DOI: 10.1002/marc.202200272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/15/2022] [Indexed: 11/09/2022]
Abstract
Physically crosslinked hydrogels have great potential for tissue engineering because of their excellent biocompatibility and easy fabrication. However, physical crosslinking points are typically weaker compared to chemical ones and therefore cannot form robust hydrogels with excellent water stability, which greatly hinder their further applications. In this work, we report a novel hydrogel with high stiffness and outstanding anti-swelling performance crosslinked by hydrophobic polymer chains entanglements. The hydrophobic polymer polyimide (PI) was mixed with the hydrophilic polymer poly(vinyl pyrrolidone) (PVP) to form crosslinking points between the chains. At the equilibrium swelling state, tensile moduli of the hydrogel can be up to 22.57 MPa (higher than most existing hydrogels) and the equilibrium water swelling ratio (ESR) can be as low as 125.0%. By decreasing the PI mass ratio, tensile moduli and ESR of the hydrogel can be tuned in a wide range from 22.57 MPa to 0.005 MPa and 125.0% to 765.6%, respectively. Using PVP/PI solutions as inks, we fabricate uniform structures and multi-material structures whose mechanical properties are close to cartilage through a direct ink writing 3D printing platform. The current work demonstrates that entangled PVP/PI hydrogels have excellent tailoring capabilities and are promising candidates for tissue engineering applications. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Suchun Ji
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, China
| | - Xiying Li
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, China
| | - Shuang Wang
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, China
| | - Hongyuan Li
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, China.,CAPT, HEDPS and IFSA Collaborative Innovation Center of MoE, Peking University, Beijing, 100871, China
| | - Huiling Duan
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, China.,CAPT, HEDPS and IFSA Collaborative Innovation Center of MoE, Peking University, Beijing, 100871, China
| | - Xin Yang
- Department of Orthopaedic, Peking University First Hospital, Beijing, 100034, China
| | - Pengyu Lv
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, China
| |
Collapse
|
108
|
Ahmed T, Liu FCF, Lu B, Lip H, Park E, Alradwan I, Liu JF, He C, Zetrini A, Zhang T, Ghavaminejad A, Rauth AM, Henderson JT, Wu XY. Advances in Nanomedicine Design: Multidisciplinary Strategies for Unmet Medical Needs. Mol Pharm 2022; 19:1722-1765. [PMID: 35587783 DOI: 10.1021/acs.molpharmaceut.2c00038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Globally, a rising burden of complex diseases takes a heavy toll on human lives and poses substantial clinical and economic challenges. This review covers nanomedicine and nanotechnology-enabled advanced drug delivery systems (DDS) designed to address various unmet medical needs. Key nanomedicine and DDSs, currently employed in the clinic to tackle some of these diseases, are discussed focusing on their versatility in diagnostics, anticancer therapy, and diabetes management. First-hand experiences from our own laboratory and the work of others are presented to provide insights into strategies to design and optimize nanomedicine- and nanotechnology-enabled DDS for enhancing therapeutic outcomes. Computational analysis is also briefly reviewed as a technology for rational design of controlled release DDS. Further explorations of DDS have illuminated the interplay of physiological barriers and their impact on DDS. It is demonstrated how such delivery systems can overcome these barriers for enhanced therapeutic efficacy and how new perspectives of next-generation DDS can be applied clinically.
Collapse
Affiliation(s)
- Taksim Ahmed
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Fuh-Ching Franky Liu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Brian Lu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - HoYin Lip
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Elliya Park
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Ibrahim Alradwan
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Jackie Fule Liu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Chunsheng He
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Abdulmottaleb Zetrini
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Tian Zhang
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Amin Ghavaminejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Andrew M Rauth
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Jeffrey T Henderson
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| |
Collapse
|
109
|
dos Santos DM, Cardoso RM, Migliorini FL, Facure MH, Mercante LA, Mattoso LH, Correa DS. Advances in 3D printed sensors for food analysis. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
110
|
Correlating Rheological Properties of a Gellan Gum-Based Bioink: A Study of the Impact of Cell Density. Polymers (Basel) 2022; 14:polym14091844. [PMID: 35567015 PMCID: PMC9102283 DOI: 10.3390/polym14091844] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/17/2022] Open
Abstract
Here, for the production of a bioink-based gellan gum, an amino derivative of this polysaccharide was mixed with a mono-functionalized aldehyde polyethyleneglycol in order to improve viscoelastic macroscopic properties and the potential processability by means of bioprinting techniques as confirmed by the printing tests. The dynamic Schiff base linkage between amino and aldehyde groups temporally modulates the rheological properties and allows a reduction of the applied pressure during extrusion followed by the recovery of gellan gum strength. Rheological properties, often related to printing resolution, were extensively investigated confirming pseudoplastic behavior and thermotropic and ionotropic responses. The success of bioprinting is related to different parameters. Among them, cell density must be carefully selected, and in order to quantify their role on printability, murine preostoblastic cells (MC3T3-E1) and human colon tumor cells (HCT-116) were chosen as cell line models. Here, we investigated the effect of their density on the bioink’s rheological properties, showing a more significant difference between cell densities for MC3T3-E1 compared to HCT-116. The results suggest the necessity of not neglecting this aspect and carrying out preliminary studies to choose the best cell densities to have the maximum viability and consequently to set the printing parameters.
Collapse
|
111
|
Green Bioprinting with Layer-by-Layer Photo-Crosslinking: A Designed Experimental Investigation on Shape Fidelity and Cell Viability of Printed Constructs. JOURNAL OF MANUFACTURING AND MATERIALS PROCESSING 2022. [DOI: 10.3390/jmmp6020045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Process variables of bioprinting (including extrusion pressure, nozzle size, and bioink composition) can affect the shape fidelity and cell viability of printed constructs. Reported studies show that increasing extrusion pressure or decreasing nozzle size would decrease cell viability in printed constructs. However, a smaller nozzle size is often necessary for printing constructs of higher shape fidelity, and a higher extrusion pressure is usually needed to extrude bioink through nozzles with a smaller diameter. Because values of printing process variables that increase shape fidelity can be detrimental to cell viability, the optimum combination of variables regarding both shape fidelity and cell viability must be determined for specific bioink compositions. This paper reports a designed experimental investigation (full factorial design with three variables and two levels) on bioprinting by applying layer-by-layer photo-crosslinking and using the alginate-methylcellulose-GelMA bioink containing algae cells. The study investigates both the main effects and interaction effects of extrusion pressure, nozzle size, and bioink composition on the shape fidelity and cell viability of printed constructs. Results show that, as extrusion pressure changed from its low level to its high level, shape fidelity and cell viability decreased. As nozzle size changed from its low level to its high level, shape fidelity decreased while cell viability increased. As bioink composition changed from its low level (with more methylcellulose content in the bioink) to its high level (with less methylcellulose content in the bioink), shape fidelity and cell viability increased.
Collapse
|
112
|
Budharaju H, Zennifer A, Sethuraman S, Paul A, Sundaramurthi D. Designer DNA biomolecules as a defined biomaterial for 3D bioprinting applications. MATERIALS HORIZONS 2022; 9:1141-1166. [PMID: 35006214 DOI: 10.1039/d1mh01632f] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
DNA has excellent features such as the presence of functional and targeted molecular recognition motifs, tailorability, multifunctionality, high-precision molecular self-assembly, hydrophilicity, and outstanding biocompatibility. Due to these remarkable features, DNA has emerged as a leading next-generation biomaterial of choice to make hydrogels by self-assembly. In recent times, novel routes for the chemical synthesis of DNA, advances in tailorable designs, and affordable production ways have made DNA as a building block material for various applications. These advanced features have made researchers continuously explore the interesting properties of pure and hybrid DNA for 3D bioprinting and other biomedical applications. This review article highlights the topical advancements in the use of DNA as an ideal bioink for the bioprinting of cell-laden three-dimensional tissue constructs for regenerative medicine applications. Various bioprinting techniques and emerging design approaches such as self-assembly, nucleotide sequence, enzymes, and production cost to use DNA as a bioink for bioprinting applications are described. In addition, various types and properties of DNA hydrogels such as stimuli responsiveness and mechanical properties are discussed. Further, recent progress in the applications of DNA in 3D bioprinting are emphasized. Finally, the current challenges and future perspectives of DNA hydrogels in 3D bioprinting and other biomedical applications are discussed.
Collapse
Affiliation(s)
- Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India.
| | - Allen Zennifer
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India.
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India.
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
- Department of Chemistry, The University of Western Ontario, London, ON N6A 5B9, Canada
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India.
| |
Collapse
|
113
|
Amirifar L, Shamloo A, Nasiri R, de Barros NR, Wang ZZ, Unluturk BD, Libanori A, Ievglevskyi O, Diltemiz SE, Sances S, Balasingham I, Seidlits SK, Ashammakhi N. Brain-on-a-chip: Recent advances in design and techniques for microfluidic models of the brain in health and disease. Biomaterials 2022; 285:121531. [DOI: 10.1016/j.biomaterials.2022.121531] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/10/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022]
|
114
|
Germain N, Dhayer M, Dekiouk S, Marchetti P. Current Advances in 3D Bioprinting for Cancer Modeling and Personalized Medicine. Int J Mol Sci 2022; 23:3432. [PMID: 35408789 PMCID: PMC8998835 DOI: 10.3390/ijms23073432] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Tumor cells evolve in a complex and heterogeneous environment composed of different cell types and an extracellular matrix. Current 2D culture methods are very limited in their ability to mimic the cancer cell environment. In recent years, various 3D models of cancer cells have been developed, notably in the form of spheroids/organoids, using scaffold or cancer-on-chip devices. However, these models have the disadvantage of not being able to precisely control the organization of multiple cell types in complex architecture and are sometimes not very reproducible in their production, and this is especially true for spheroids. Three-dimensional bioprinting can produce complex, multi-cellular, and reproducible constructs in which the matrix composition and rigidity can be adapted locally or globally to the tumor model studied. For these reasons, 3D bioprinting seems to be the technique of choice to mimic the tumor microenvironment in vivo as closely as possible. In this review, we discuss different 3D-bioprinting technologies, including bioinks and crosslinkers that can be used for in vitro cancer models and the techniques used to study cells grown in hydrogels; finally, we provide some applications of bioprinted cancer models.
Collapse
Affiliation(s)
- Nicolas Germain
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| | - Melanie Dhayer
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Salim Dekiouk
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Philippe Marchetti
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| |
Collapse
|
115
|
Iglesias-Mejuto A, García-González CA. 3D-Printed, Dual Crosslinked and Sterile Aerogel Scaffolds for Bone Tissue Engineering. Polymers (Basel) 2022; 14:polym14061211. [PMID: 35335542 PMCID: PMC8951756 DOI: 10.3390/polym14061211] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
The fabrication of bioactive three-dimensional (3D) hydrogel scaffolds from biocompatible materials with a complex inner structure (mesoporous and macroporous) and highly interconnected porosity is crucial for bone tissue engineering (BTE). 3D-printing technology combined with aerogel processing allows the fabrication of functional nanostructured scaffolds from polysaccharides for BTE with personalized geometry, porosity and composition. However, these aerogels are usually fragile, with fast biodegradation rates in biological aqueous fluids, and they lack the sterility required for clinical practice. In this work, reinforced alginate-hydroxyapatite (HA) aerogel scaffolds for BTE applications were obtained by a dual strategy that combines extrusion-based 3D-printing and supercritical CO2 gel drying with an extra crosslinking step. Gel ageing in CaCl2 solutions and glutaraldehyde (GA) chemical crosslinking of aerogels were performed as intermediate and post-processing reinforcement strategies to achieve highly crosslinked aerogel scaffolds. Nitrogen adsorption–desorption (BET) and SEM analyses were performed to assess the textural parameters of the resulting alginate-HA aerogel scaffolds. The biological evaluation of the aerogel scaffolds was performed regarding cell viability, hemolytic activity and bioactivity for BTE. The impact of scCO2-based post-sterilization treatment on scaffold properties was also assessed. The obtained aerogels were dual porous, bio- and hemocompatible, as well as endowed with high bioactivity that is dependent on the HA content. This work is a step forward towards the optimization of the physicochemical performance of advanced biomaterials and their sterilization.
Collapse
|
116
|
Zhu H, Monavari M, Zheng K, Distler T, Ouyang L, Heid S, Jin Z, He J, Li D, Boccaccini AR. 3D Bioprinting of Multifunctional Dynamic Nanocomposite Bioinks Incorporating Cu-Doped Mesoporous Bioactive Glass Nanoparticles for Bone Tissue Engineering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104996. [PMID: 35102718 DOI: 10.1002/smll.202104996] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/28/2021] [Indexed: 06/14/2023]
Abstract
Bioprinting has seen significant progress in recent years for the fabrication of bionic tissues with high complexity. However, it remains challenging to develop cell-laden bioinks exhibiting superior physiochemical properties and bio-functionality. In this study, a multifunctional nanocomposite bioink is developed based on amine-functionalized copper (Cu)-doped mesoporous bioactive glass nanoparticles (ACuMBGNs) and a hydrogel formulation relying on dynamic covalent chemistry composed of alginate dialdehyde (oxidized alginate) and gelatin, with favorable rheological properties, improved shape fidelity, and structural stability for extrusion-based bioprinting. The reversible dynamic microenvironment in combination with the impact of cell-adhesive ligands introduced by aminated particles enables the rapid spreading (within 3 days) and high survival (>90%) of embedded human osteosarcoma cells and immortalized mouse bone marrow-derived stroma cells. Osteogenic differentiation of primary mouse bone marrow stromal stem cells (BMSCs) and angiogenesis are promoted in the bioprinted alginate dialdehyde-gelatin (ADA-GEL or AG)-ACuMBGN scaffolds without additional growth factors in vitro, which is likely due to ion stimulation from the incorporated nanoparticles and possibly due to cell mechanosensing in the dynamic matrix. In conclusion, it is envisioned that these nanocomposite bioinks can serve as promising platforms for bioprinting complex 3D matrix environments providing superior physiochemical and biological performance for bone tissue engineering.
Collapse
Affiliation(s)
- Hui Zhu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Mahshid Monavari
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Kai Zheng
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Thomas Distler
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Liliang Ouyang
- Department of Mechanical Engineering, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Tsinghua University, Beijing, 100084, P. R. China
| | - Susanne Heid
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Zhaorui Jin
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| | - Dichen Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| | - Aldo R Boccaccini
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| |
Collapse
|
117
|
Kurian AG, Singh RK, Patel KD, Lee JH, Kim HW. Multifunctional GelMA platforms with nanomaterials for advanced tissue therapeutics. Bioact Mater 2022; 8:267-295. [PMID: 34541401 PMCID: PMC8424393 DOI: 10.1016/j.bioactmat.2021.06.027] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Polymeric hydrogels are fascinating platforms as 3D scaffolds for tissue repair and delivery systems of therapeutic molecules and cells. Among others, methacrylated gelatin (GelMA) has become a representative hydrogel formulation, finding various biomedical applications. Recent efforts on GelMA-based hydrogels have been devoted to combining them with bioactive and functional nanomaterials, aiming to provide enhanced physicochemical and biological properties to GelMA. The benefits of this approach are multiple: i) reinforcing mechanical properties, ii) modulating viscoelastic property to allow 3D printability of bio-inks, iii) rendering electrical/magnetic property to produce electro-/magneto-active hydrogels for the repair of specific tissues (e.g., muscle, nerve), iv) providing stimuli-responsiveness to actively deliver therapeutic molecules, and v) endowing therapeutic capacity in tissue repair process (e.g., antioxidant effects). The nanomaterial-combined GelMA systems have shown significantly enhanced and extraordinary behaviors in various tissues (bone, skin, cardiac, and nerve) that are rarely observable with GelMA. Here we systematically review these recent efforts in nanomaterials-combined GelMA hydrogels that are considered as next-generation multifunctional platforms for tissue therapeutics. The approaches used in GelMA can also apply to other existing polymeric hydrogel systems.
Collapse
Affiliation(s)
- Amal George Kurian
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Rajendra K. Singh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Kapil D. Patel
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, London, WC1X8LD, UK
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
118
|
Remaggi G, Catanzano O, Quaglia F, Elviri L. Alginate Self-Crosslinking Ink for 3D Extrusion-Based Cryoprinting and Application for Epirubicin-HCl Delivery on MCF-7 Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030882. [PMID: 35164146 PMCID: PMC8839018 DOI: 10.3390/molecules27030882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 11/17/2022]
Abstract
3D-printed hydrogels are particularly advantageous as drug-delivery platforms but their loading with water-soluble active compounds remains a challenge requiring the development of innovative inks. Here, we propose a new 3D extrusion-based approach that, by exploiting the internal gelation of the alginate, avoids the post-printing crosslinking process and allows the loading of epirubicin-HCl (EPI). The critical combinations of alginate, calcium carbonate and d-glucono-δ-lactone (GDL) combined with the scaffold production parameters (extrusion time, temperature, and curing time) were evaluated and discussed. The internal gelation in tandem with 3D extrusion allowed the preparation of alginate hydrogels with a complex shape and good handling properties. The dispersion of epirubicin-HCl in the hydrogel matrix confirmed the potential of this self-crosslinking alginate-based ink for the preparation of 3D-printed drug-delivery platforms. Drug release from 3D-printed hydrogels was monitored, and the cytotoxic activity was tested against MCF-7 cells. Finally, the change in the expression pattern of anti-apoptotic, pro-apoptotic, and autophagy protein markers was monitored by liquid-chromatography tandem-mass-spectrometry after exposure of MCF-7 to the EPI-loaded hydrogels.
Collapse
Affiliation(s)
- Giulia Remaggi
- Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy;
| | - Ovidio Catanzano
- Institute for Polymers, Composites and Biomaterials (IPCB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, NA, Italy;
| | - Fabiana Quaglia
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy;
| | - Lisa Elviri
- Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy;
- Correspondence:
| |
Collapse
|
119
|
Behan K, Dufour A, Garcia O, Kelly D. Methacrylated Cartilage ECM-Based Hydrogels as Injectables and Bioinks for Cartilage Tissue Engineering. Biomolecules 2022; 12:biom12020216. [PMID: 35204718 PMCID: PMC8961582 DOI: 10.3390/biom12020216] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/22/2022] Open
Abstract
Articular cartilage (AC) possesses a limited healing potential, meaning that untreated focal joint defects typically progress, leading to the development of degenerative diseases such as osteoarthritis. Several clinical strategies exist that aim to regenerate AC; however, recapitulation of a fully functional, load-bearing tissue remains a significant challenge. This can be attributed, at least in part, to a paucity of biomaterials that truly mimic the native tissue and provide appropriate cues to direct its regeneration. The main structural component of articular cartilage, type II collagen, does not readily gelate at body temperature, challenging the development of cartilage extracellular matrix (cECM)-derived injectable hydrogels and bioinks for AC tissue engineering and bioprinting applications. Here, we describe the development and rheological characterisation of a methacrylated cartilage ECM-based hydrogel/bioink (cECM-MA), which could be photocrosslinked when exposed to ultraviolet (UV) light. Functionalisation of the collagen backbone with methacryloyl groups had a negligible effect on triple helix stability, as demonstrated by circular dichroism spectroscopy. These cECM-MA bioinks demonstrated shear-thinning properties and could be loaded with bone marrow mesenchymal stem cells (BM-MSCs), micro-extruded to generate self-supporting 3D constructs of predefined size and shape, and then photocrosslinked using UV light. Analysis of the cell-laden constructs showed that the BM-MSCs were viable post-printing and underwent chondrogenesis in vitro, generating a tissue rich in sulphated glycosaminoglycans and collagens. These results support the use of methacrylated, tissue-specific ECM-derived hydrogels as bioinks for 3D bioprinting and/or as injectables for cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Kevin Behan
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (K.B.); (A.D.)
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Alexandre Dufour
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (K.B.); (A.D.)
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Orquidea Garcia
- Johnson & Johnson Services, Inc., 31 Technology Drive, Irvine, CA 92618, USA;
| | - Daniel Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (K.B.); (A.D.)
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), CRANN, Naughton Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Department of Anatomy, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- Correspondence:
| |
Collapse
|
120
|
Mueller E, Poulin I, Bodnaryk WJ, Hoare T. Click Chemistry Hydrogels for Extrusion Bioprinting: Progress, Challenges, and Opportunities. Biomacromolecules 2022; 23:619-640. [PMID: 34989569 DOI: 10.1021/acs.biomac.1c01105] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The emergence of 3D bioprinting has allowed a variety of hydrogel-based "bioinks" to be printed in the presence of cells to create precisely defined cell-loaded 3D scaffolds in a single step for advancing tissue engineering and/or regenerative medicine. While existing bioinks based primarily on ionic cross-linking, photo-cross-linking, or thermogelation have significantly advanced the field, they offer technical limitations in terms of the mechanics, degradation rates, and the cell viabilities achievable with the printed scaffolds, particularly in terms of aiming to match the wide range of mechanics and cellular microenvironments. Click chemistry offers an appealing solution to this challenge given that proper selection of the chemistry can enable precise tuning of both the gelation rate and the degradation rate, both key to successful tissue regeneration; simultaneously, the often bio-orthogonal nature of click chemistry is beneficial to maintain high cell viabilities within the scaffolds. However, to date, relatively few examples of 3D-printed click chemistry hydrogels have been reported, mostly due to the technical challenges of controlling mixing during the printing process to generate high-fidelity prints without clogging the printer. This review aims to showcase existing cross-linking modalities, characterize the advantages and disadvantages of different click chemistries reported, highlight current examples of click chemistry hydrogel bioinks, and discuss the design of mixing strategies to enable effective 3D extrusion bioprinting of click hydrogels.
Collapse
Affiliation(s)
- Eva Mueller
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L7, Canada
| | - Isabelle Poulin
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L7, Canada
| | - William James Bodnaryk
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L7, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L7, Canada
| |
Collapse
|
121
|
Emerging trends and prospects of electroconductive bioinks for cell-laden and functional 3D bioprinting. Biodes Manuf 2022. [DOI: 10.1007/s42242-021-00169-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
122
|
Xu L, Ye Q, Xie J, Yang J, Jiang W, Yuan H, Li J. An injectable gellan gum-based hydrogel that inhibits Staphylococcus aureus for infected bone defect repair. J Mater Chem B 2022; 10:282-292. [PMID: 34908091 DOI: 10.1039/d1tb02230j] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The treatment of infected bone defects in complex anatomical structures, such as oral and maxillofacial structures, remains an intractable clinical challenge. Therefore, advanced biomaterials that have excellent anti-infection activity and allow convenient delivery are needed. We fabricated an innovative injectable gellan gum (GG)-based hydrogel loaded with nanohydroxyapatite particles and chlorhexidine (nHA/CHX). The hydrogel has a porous morphology, suitable swelling ratio, and good biocompatibility. It exerts strong antibacterial activity against Staphylococcus aureus growth and biofilm formation in vitro. We successfully established an infected calvarial defect rat model. Bacterial colony numbers were significantly lower in tissues surrounding the bone in rats of the GG/nHA/CHX group after debride surgery and hydrogel implantation in the defect regions than in rats of the blank group. Rats in the GG/nHA/CHX group exhibited significantly increased new bone formation compared to those in the blank group at 4 and 8 weeks. These findings indicate that gellan gum-based hydrogel with nHA/CHX can accelerate the repair of infected bone defects.
Collapse
Affiliation(s)
- Laijun Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
- Department of Operative Dentistry and Endodontics, Xiangya School of Stomatology, Xiangya Stomatological Hospital, Central South University, Changsha, 410008, China
| | - Qing Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Jing Xie
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Jiaojiao Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Wentao Jiang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Province Key Laboratory of Stomatology, Guangzhou, 510060, China
| | - He Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Jiyao Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
123
|
Li M, Sun D, Zhang J, Wang Y, Wei Q, Wang Y. Application and development of 3D bioprinting in cartilage tissue engineering. Biomater Sci 2022; 10:5430-5458. [DOI: 10.1039/d2bm00709f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bioprinting technology can build complex tissue structures and has the potential to fabricate engineered cartilage with bionic structures for achieving cartilage defect repair/regeneration.
Collapse
Affiliation(s)
- Mingyang Li
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Daocen Sun
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Juan Zhang
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yanmei Wang
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Qinghua Wei
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yanen Wang
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
124
|
Cho KW, Sunwoo SH, Hong YJ, Koo JH, Kim JH, Baik S, Hyeon T, Kim DH. Soft Bioelectronics Based on Nanomaterials. Chem Rev 2021; 122:5068-5143. [PMID: 34962131 DOI: 10.1021/acs.chemrev.1c00531] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent advances in nanostructured materials and unconventional device designs have transformed the bioelectronics from a rigid and bulky form into a soft and ultrathin form and brought enormous advantages to the bioelectronics. For example, mechanical deformability of the soft bioelectronics and thus its conformal contact onto soft curved organs such as brain, heart, and skin have allowed researchers to measure high-quality biosignals, deliver real-time feedback treatments, and lower long-term side-effects in vivo. Here, we review various materials, fabrication methods, and device strategies for flexible and stretchable electronics, especially focusing on soft biointegrated electronics using nanomaterials and their composites. First, we summarize top-down material processing and bottom-up synthesis methods of various nanomaterials. Next, we discuss state-of-the-art technologies for intrinsically stretchable nanocomposites composed of nanostructured materials incorporated in elastomers or hydrogels. We also briefly discuss unconventional device design strategies for soft bioelectronics. Then individual device components for soft bioelectronics, such as biosensing, data storage, display, therapeutic stimulation, and power supply devices, are introduced. Afterward, representative application examples of the soft bioelectronics are described. A brief summary with a discussion on remaining challenges concludes the review.
Collapse
Affiliation(s)
- Kyoung Won Cho
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea.,Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung-Hyuk Sunwoo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Yongseok Joseph Hong
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Ja Hoon Koo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Jeong Hyun Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Seungmin Baik
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea.,Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dae-Hyeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea.,Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.,Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
125
|
Gao Q, Kim BS, Gao G. Advanced Strategies for 3D Bioprinting of Tissue and Organ Analogs Using Alginate Hydrogel Bioinks. Mar Drugs 2021; 19:708. [PMID: 34940707 PMCID: PMC8708555 DOI: 10.3390/md19120708] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/12/2021] [Accepted: 12/12/2021] [Indexed: 12/15/2022] Open
Abstract
Alginate is a natural polysaccharide that typically originates from various species of algae. Due to its low cost, good biocompatibility, and rapid ionic gelation, the alginate hydrogel has become a good option of bioink source for 3D bioprinting. However, the lack of cell adhesive moieties, erratic biodegradability, and poor printability are the critical limitations of alginate hydrogel bioink. This review discusses the pivotal properties of alginate hydrogel as a bioink for 3D bioprinting technologies. Afterward, a variety of advanced material formulations and biofabrication strategies that have recently been developed to overcome the drawbacks of alginate hydrogel bioink will be focused on. In addition, the applications of these advanced solutions for 3D bioprinting of tissue/organ mimicries such as regenerative implants and in vitro tissue models using alginate-based bioink will be systematically summarized.
Collapse
Affiliation(s)
- Qiqi Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China;
| | - Byoung-Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 626841, Kyungnam, Korea;
| | - Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China;
- Department of Medical Technology, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China
| |
Collapse
|
126
|
Tan Z, Bilal M, Raza A, Cui J, Ashraf SS, Iqbal HMN. Expanding the Biocatalytic Scope of Enzyme-Loaded Polymeric Hydrogels. Gels 2021; 7:gels7040194. [PMID: 34842692 PMCID: PMC8628689 DOI: 10.3390/gels7040194] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 02/05/2023] Open
Abstract
In recent years, polymeric hydrogels have appeared promising matrices for enzyme immobilization to design, signify and expand bio-catalysis engineering. Therefore, the development and deployment of polymeric supports in the form of hydrogels and other robust geometries are continuously growing to green the twenty-first-century bio-catalysis. Furthermore, adequately fabricated polymeric hydrogel materials offer numerous advantages that shield pristine enzymes from denaturation under harsh reaction environments. For instance, cross-linking modulation of hydrogels, distinct rheological behavior, tunable surface entities along with elasticity and mesh size, larger surface-volume area, and hydrogels' mechanical cushioning attributes are of supreme interest makes them the ideal candidate for enzyme immobilization. Furthermore, suitable coordination of polymeric hydrogels with requisite enzyme fraction enables pronounced loading, elevated biocatalytic activity, and exceptional stability. Additionally, the unique catalytic harmony of enzyme-loaded polymeric hydrogels offers numerous applications, such as hydrogels as immobilization matrix, bio-catalysis, sensing, detection and monitoring, tissue engineering, wound healing, and drug delivery applications. In this review, we spotlight the applied perspective of enzyme-loaded polymeric hydrogels with recent and relevant examples. The work also signifies the combined use of multienzyme systems and the future directions that should be attempted in this field.
Collapse
Affiliation(s)
- Zhongbiao Tan
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China;
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China;
- Correspondence: (M.B.); (H.M.N.I.)
| | - Ali Raza
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China;
| | - Jiandong Cui
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, No 29, 13th, Avenue, Tianjin Economic and Technological Development Area (TEDA), Tianjin 300457, China;
| | - Syed Salman Ashraf
- Department of Biology, College of Arts and Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates;
- Center for Biotechnology (BTC), Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Hafiz M. N. Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
- Correspondence: (M.B.); (H.M.N.I.)
| |
Collapse
|
127
|
Functional role of crosslinking in alginate scaffold for drug delivery and tissue engineering: A review. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2021.110807] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
128
|
Rastin H, Mansouri N, Tung TT, Hassan K, Mazinani A, Ramezanpour M, Yap PL, Yu L, Vreugde S, Losic D. Converging 2D Nanomaterials and 3D Bioprinting Technology: State-of-the-Art, Challenges, and Potential Outlook in Biomedical Applications. Adv Healthc Mater 2021; 10:e2101439. [PMID: 34468088 DOI: 10.1002/adhm.202101439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Indexed: 12/17/2022]
Abstract
The development of next-generation of bioinks aims to fabricate anatomical size 3D scaffold with high printability and biocompatibility. Along with the progress in 3D bioprinting, 2D nanomaterials (2D NMs) prove to be emerging frontiers in the development of advanced materials owing to their extraordinary properties. Harnessing the properties of 2D NMs in 3D bioprinting technologies can revolutionize the development of bioinks by endowing new functionalities to the current bioinks. First the main contributions of 2D NMS in 3D bioprinting technologies are categorized here into six main classes: 1) reinforcement effect, 2) delivery of bioactive molecules, 3) improved electrical conductivity, 4) enhanced tissue formation, 5) photothermal effect, 6) and stronger antibacterial properties. Next, the recent advances in the use of each certain 2D NMs (1) graphene, 2) nanosilicate, 3) black phosphorus, 4) MXene, 5) transition metal dichalcogenides, 6) hexagonal boron nitride, and 7) metal-organic frameworks) in 3D bioprinting technology are critically summarized and evaluated thoroughly. Third, the role of physicochemical properties of 2D NMSs on their cytotoxicity is uncovered, with several representative examples of each studied 2D NMs. Finally, current challenges, opportunities, and outlook for the development of nanocomposite bioinks are discussed thoroughly.
Collapse
Affiliation(s)
- Hadi Rastin
- School of Chemical Engineering and Advanced Materials The University of Adelaide South Australia 5005 Australia
- ARC Research Hub for Graphene Enabled Industry Transformation The University of Adelaide South Australia 5005 Australia
| | - Negar Mansouri
- School of Chemical Engineering and Advanced Materials The University of Adelaide South Australia 5005 Australia
- School of Electrical and Electronic Engineering The University of Adelaide South Australia 5005 Australia
| | - Tran Thanh Tung
- School of Chemical Engineering and Advanced Materials The University of Adelaide South Australia 5005 Australia
- ARC Research Hub for Graphene Enabled Industry Transformation The University of Adelaide South Australia 5005 Australia
| | - Kamrul Hassan
- School of Chemical Engineering and Advanced Materials The University of Adelaide South Australia 5005 Australia
- ARC Research Hub for Graphene Enabled Industry Transformation The University of Adelaide South Australia 5005 Australia
| | - Arash Mazinani
- School of Chemical Engineering and Advanced Materials The University of Adelaide South Australia 5005 Australia
- ARC Research Hub for Graphene Enabled Industry Transformation The University of Adelaide South Australia 5005 Australia
| | - Mahnaz Ramezanpour
- Department of Surgery‐Otolaryngology Head and Neck Surgery The University of Adelaide Woodville South 5011 Australia
| | - Pei Lay Yap
- School of Chemical Engineering and Advanced Materials The University of Adelaide South Australia 5005 Australia
- ARC Research Hub for Graphene Enabled Industry Transformation The University of Adelaide South Australia 5005 Australia
| | - Le Yu
- School of Chemical Engineering and Advanced Materials The University of Adelaide South Australia 5005 Australia
- ARC Research Hub for Graphene Enabled Industry Transformation The University of Adelaide South Australia 5005 Australia
| | - Sarah Vreugde
- Department of Surgery‐Otolaryngology Head and Neck Surgery The University of Adelaide Woodville South 5011 Australia
| | - Dusan Losic
- School of Chemical Engineering and Advanced Materials The University of Adelaide South Australia 5005 Australia
- ARC Research Hub for Graphene Enabled Industry Transformation The University of Adelaide South Australia 5005 Australia
| |
Collapse
|
129
|
Fang Y, Shi L, Duan Z, Rohani S. Hyaluronic acid hydrogels, as a biological macromolecule-based platform for stem cells delivery and their fate control: A review. Int J Biol Macromol 2021; 189:554-566. [PMID: 34437920 DOI: 10.1016/j.ijbiomac.2021.08.140] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 11/27/2022]
Abstract
Stem cell-based therapies offer numerous potentials to repair damaged or defective organs. The therapeutic outcomes of human studies, however, fall far short from what is expected. Enhancing stem cells local density and longevity would possibly maximize their healing potential. One promising strategy is to administer stem cells via injectable hydrogels. However, stem cells differentiation process is a delicate matter which is easily affected by various factors such as their interaction with their surrounding materials. Among various biomaterial options for hydrogels' production, hyaluronic acid (HA) has shown great promise. HA is a naturally occurring biological macromolecule, a polysaccharide of large molecular weight which is involved in cell proliferation, cell migration, angiogenesis, fetal development, and tissue function. In the current study we will discuss the applications, prospects, and challenges of HA-based hydrogels in stem cell delivery and fate control.
Collapse
Affiliation(s)
- Yu Fang
- Henan Provincial Engineering and Technology Research Center for Precise Synthesis of Fluorine-Containing Drugs, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China; Key Laboratory of New Opto-Electronic Functional Materials of Henan Province, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China.
| | - Lele Shi
- Henan Provincial Engineering and Technology Research Center for Precise Synthesis of Fluorine-Containing Drugs, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China; Key Laboratory of New Opto-Electronic Functional Materials of Henan Province, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China
| | - Zhiwei Duan
- Henan Provincial Engineering and Technology Research Center for Precise Synthesis of Fluorine-Containing Drugs, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China; Key Laboratory of New Opto-Electronic Functional Materials of Henan Province, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China
| | - Saeed Rohani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
130
|
McGivern S, Boutouil H, Al-Kharusi G, Little S, Dunne NJ, Levingstone TJ. Translational Application of 3D Bioprinting for Cartilage Tissue Engineering. Bioengineering (Basel) 2021; 8:144. [PMID: 34677217 PMCID: PMC8533558 DOI: 10.3390/bioengineering8100144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/07/2021] [Accepted: 10/10/2021] [Indexed: 12/16/2022] Open
Abstract
Cartilage is an avascular tissue with extremely limited self-regeneration capabilities. At present, there are no existing treatments that effectively stop the deterioration of cartilage or reverse its effects; current treatments merely relieve its symptoms and surgical intervention is required when the condition aggravates. Thus, cartilage damage remains an ongoing challenge in orthopaedics with an urgent need for improved treatment options. In recent years, major advances have been made in the development of three-dimensional (3D) bioprinted constructs for cartilage repair applications. 3D bioprinting is an evolutionary additive manufacturing technique that enables the precisely controlled deposition of a combination of biomaterials, cells, and bioactive molecules, collectively known as bioink, layer-by-layer to produce constructs that simulate the structure and function of native cartilage tissue. This review provides an insight into the current developments in 3D bioprinting for cartilage tissue engineering. The bioink and construct properties required for successful application in cartilage repair applications are highlighted. Furthermore, the potential for translation of 3D bioprinted constructs to the clinic is discussed. Overall, 3D bioprinting demonstrates great potential as a novel technique for the fabrication of tissue engineered constructs for cartilage regeneration, with distinct advantages over conventional techniques.
Collapse
Affiliation(s)
- Sophie McGivern
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, D09 NA55 Dublin, Ireland; (S.M.); (H.B.); (G.A.-K.); (N.J.D.)
| | - Halima Boutouil
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, D09 NA55 Dublin, Ireland; (S.M.); (H.B.); (G.A.-K.); (N.J.D.)
- Centre for Medical Engineering Research (MEDeng), Dublin City University, D09 NA55 Dublin, Ireland
| | - Ghayadah Al-Kharusi
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, D09 NA55 Dublin, Ireland; (S.M.); (H.B.); (G.A.-K.); (N.J.D.)
- Centre for Medical Engineering Research (MEDeng), Dublin City University, D09 NA55 Dublin, Ireland
| | - Suzanne Little
- Insight SFI Research Centre for Data Analytics, Dublin City University, D09 NA55 Dublin, Ireland;
| | - Nicholas J. Dunne
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, D09 NA55 Dublin, Ireland; (S.M.); (H.B.); (G.A.-K.); (N.J.D.)
- Centre for Medical Engineering Research (MEDeng), Dublin City University, D09 NA55 Dublin, Ireland
- Advanced Processing Technology Research Centre, Dublin City University, D09 NA55 Dublin, Ireland
- Biodesign Europe, Dublin City University, D09 NA55 Dublin, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 PN40 Dublin, Ireland
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Tanya J. Levingstone
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, D09 NA55 Dublin, Ireland; (S.M.); (H.B.); (G.A.-K.); (N.J.D.)
- Centre for Medical Engineering Research (MEDeng), Dublin City University, D09 NA55 Dublin, Ireland
- Advanced Processing Technology Research Centre, Dublin City University, D09 NA55 Dublin, Ireland
- Biodesign Europe, Dublin City University, D09 NA55 Dublin, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| |
Collapse
|
131
|
Honaryar H, LaNasa JA, Lloyd EC, Hickey RJ, Niroobakhsh Z. Fabricating Robust Constructs with Internal Phase Nanostructures via Liquid-in-Liquid 3D Printing. Macromol Rapid Commun 2021; 42:e2100445. [PMID: 34569682 DOI: 10.1002/marc.202100445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/21/2021] [Indexed: 12/12/2022]
Abstract
The ability to print soft materials into predefined architectures with programmable nanostructures and mechanical properties is a necessary requirement for creating synthetic biomaterials that mimic living tissues. However, the low viscosity of common materials and lack of required mechanical properties in the final product present an obstacle to the use of traditional additive manufacturing approaches. Here, a new liquid-in-liquid 3D printing approach is used to successfully fabricate constructs with internal nanostructures using in situ self-assembly during the extrusion of an aqueous solution containing surfactant and photocurable polymer into a stabilizing polar oil bath. Subsequent photopolymerization preserves the nanostructures created due to surfactant self-assembly at the immiscible liquid-liquid interface, which is confirmed by small-angle X-ray scattering. Mechanical properties of the photopolymerized prints are shown to be tunable based on constituent components of the aqueous solution. The reported 3D printing approach expands the range of low-viscosity materials that can be used in 3D printing, and enables robust constructs production with internal nanostructures and spatially defined features. The reported approach has broad applications in regenerative medicine by providing a platform to print self-assembling biomaterials into complex tissue mimics where internal supramolecular structures and their functionality control biological processes, similar to natural extracellular matrices.
Collapse
Affiliation(s)
- Houman Honaryar
- Department of Civil & Mechanical Engineering, University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Jacob A LaNasa
- Department of Materials Science & Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Elisabeth C Lloyd
- Department of Materials Science & Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Robert J Hickey
- Department of Materials Science & Engineering, Pennsylvania State University, University Park, PA, 16802, USA.,Materials Research Institute, Pennsylvania State University, University Park, PA, 16802, USA
| | - Zahra Niroobakhsh
- Department of Civil & Mechanical Engineering, University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| |
Collapse
|
132
|
Uppuluri VNVA, Thukani Sathanantham S, Bhimavarapu SK, Elumalai L. Polymeric Hydrogel Scaffolds: Skin Tissue Engineering and Regeneration. Adv Pharm Bull 2021; 12:437-448. [PMID: 35935050 PMCID: PMC9348527 DOI: 10.34172/apb.2022.069] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/22/2021] [Accepted: 09/13/2021] [Indexed: 11/09/2022] Open
Abstract
Tissue engineering is a novel regenerative approach in the medicinal field that promises the regeneration of damaged tissues. Moreover, tissue engineering involves synthetic and natural biomaterials that facilitate tissue or organ growth outside the body. Not surprisingly, the demand for polymer-based therapeutical approaches in skin tissue defects has increased at an effective rate, despite the pressing clinical need. Among the 3D scaffolds for tissue engineering and regeneration approaches, hydrogel scaffolds have shown significant importance for their use as 3D cross-linked scaffolds in skin tissue regeneration due to their ideal moisture retention property and porosity biocompatibility, biodegradable, and biomimetic characteristics. In this review, we demonstrated the choice of ideal biomaterials to fabricate the novel hydrogel scaffolds for skin tissue engineering. After a short introduction to the bioactive and drug-loaded polymeric hydrogels, the discussion turns to fabrication and characterisation techniques of the polymeric hydrogel scaffolds. In conclusion, we discuss the excellent wound healing potential of stem cell-loaded hydrogels and Nano-based approaches to designing hydrogel scaffolds for skin tissue engineering.
Collapse
Affiliation(s)
- Varuna Naga Venkata Arjun Uppuluri
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology & Advanced Studies (VISTAS), Chennai, 600 117, Tamil Nadu, India
| | - Shanmugarajan Thukani Sathanantham
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology & Advanced Studies (VISTAS), Chennai, 600 117, Tamil Nadu, India
| | - Sai Krishna Bhimavarapu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology & Advanced Studies (VISTAS), Chennai, 600 117, Tamil Nadu, India
| | - Lokesh Elumalai
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology & Advanced Studies (VISTAS), Chennai, 600 117, Tamil Nadu, India
| |
Collapse
|
133
|
Veiga A, Silva IV, Duarte MM, Oliveira AL. Current Trends on Protein Driven Bioinks for 3D Printing. Pharmaceutics 2021; 13:1444. [PMID: 34575521 PMCID: PMC8471984 DOI: 10.3390/pharmaceutics13091444] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023] Open
Abstract
In the last decade, three-dimensional (3D) extrusion bioprinting has been on the top trend for innovative technologies in the field of biomedical engineering. In particular, protein-based bioinks such as collagen, gelatin, silk fibroin, elastic, fibrin and protein complexes based on decellularized extracellular matrix (dECM) are receiving increasing attention. This current interest is the result of protein's tunable properties, biocompatibility, environmentally friendly nature and possibility to provide cells with the adequate cues, mimicking the extracellular matrix's function. In this review we describe the most relevant stages of the development of a protein-driven bioink. The most popular formulations, molecular weights and extraction methods are covered. The different crosslinking methods used in protein bioinks, the formulation with other polymeric systems or molecules of interest as well as the bioprinting settings are herein highlighted. The cell embedding procedures, the in vitro, in vivo, in situ studies and final applications are also discussed. Finally, we approach the development and optimization of bioinks from a sequential perspective, discussing the relevance of each parameter during the pre-processing, processing, and post-processing stages of technological development. Through this approach the present review expects to provide, in a sequential manner, helpful methodological guidelines for the development of novel bioinks.
Collapse
Affiliation(s)
- Anabela Veiga
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, 4099-002 Porto, Portugal
| | - Inês V. Silva
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
| | - Marta M. Duarte
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
| | - Ana L. Oliveira
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
| |
Collapse
|
134
|
Gao G, Ahn M, Cho WW, Kim BS, Cho DW. 3D Printing of Pharmaceutical Application: Drug Screening and Drug Delivery. Pharmaceutics 2021; 13:1373. [PMID: 34575448 PMCID: PMC8465948 DOI: 10.3390/pharmaceutics13091373] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/20/2021] [Accepted: 08/29/2021] [Indexed: 12/22/2022] Open
Abstract
Advances in three-dimensional (3D) printing techniques and the development of tailored biomaterials have facilitated the precise fabrication of biological components and complex 3D geometrics over the past few decades. Moreover, the notable growth of 3D printing has facilitated pharmaceutical applications, enabling the development of customized drug screening and drug delivery systems for individual patients, breaking away from conventional approaches that primarily rely on transgenic animal experiments and mass production. This review provides an extensive overview of 3D printing research applied to drug screening and drug delivery systems that represent pharmaceutical applications. We classify several elements required by each application for advanced pharmaceutical techniques and briefly describe state-of-the-art 3D printing technology consisting of cells, bioinks, and printing strategies that satisfy requirements. Furthermore, we discuss the limitations of traditional approaches by providing concrete examples of drug screening (organoid, organ-on-a-chip, and tissue/organ equivalent) and drug delivery systems (oral/vaginal/rectal and transdermal/surgical drug delivery), followed by the introduction of recent pharmaceutical investigations using 3D printing-based strategies to overcome these challenges.
Collapse
Affiliation(s)
- Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China;
| | - Minjun Ahn
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| | - Won-Woo Cho
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| | - Byoung-Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Kyungbuk, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| |
Collapse
|
135
|
Bioprinting of Organ-on-Chip Systems: A Literature Review from a Manufacturing Perspective. JOURNAL OF MANUFACTURING AND MATERIALS PROCESSING 2021. [DOI: 10.3390/jmmp5030091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review discusses the reported studies investigating the use of bioprinting to develop functional organ-on-chip systems from a manufacturing perspective. These organ-on-chip systems model the liver, kidney, heart, lung, gut, bone, vessel, and tumors to demonstrate the viability of bioprinted organ-on-chip systems for disease modeling and drug screening. In addition, the paper highlights the challenges involved in using bioprinting techniques for organ-on-chip system fabrications and suggests future research directions. Based on the reviewed studies, it is concluded that bioprinting can be applied for the automated and assembly-free fabrication of organ-on chip systems. These bioprinted organ-on-chip systems can help in the modeling of several different diseases and can thereby expedite drug discovery by providing an efficient platform for drug screening in the preclinical phase of drug development processes.
Collapse
|
136
|
Huang Y, Zhou Z, Hu Y, He N, Li J, Han X, Zhao G, Liu H. Modified mannan for 3D bioprinting: a potential novel bioink for tissue engineering. Biomed Mater 2021; 16. [PMID: 34348252 DOI: 10.1088/1748-605x/ac1ab4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/04/2021] [Indexed: 02/05/2023]
Abstract
3D bioprinting technology displays many advantages for tissue engineering applications, but its utilization is limited by veryfew bioinks available for biofabrication. In this study, a novel type of bioink, which includes three methacryloyl modifiedmannans, was introduced to 3D bioprinting for tissue engineering applications. Yeast mannan (YM) was modified by reactingwith methacrylate anhydride (MA) at different concentrations, and three YM derived bioinks were obtained, which weretermed as YM-MA-1, YM-MA-2 and YM-MA-3 and were distinguished with different adjusted methacrylation degrees. TheYM derived bioink displayed an advantage that the mechanical properties of its photo-cured hydrogels can be enhanced withits methacrylation degree. Hence, YM derived bioinks are fitted for the mechanical requirements of most soft tissueengineering, including cartilage tissue engineering. By selecting chondrocytes as the testing cells, well cytocompatibility of YM-MA-1, YM-MA-2 had been confirmed by CCK-8 method. Following photo-crosslinking and implantation into SD rats for 4 weeks, thein vivobiocompatibility of the YM-MA-2 hydrogel is acceptable for tissue engineering applications. Hence, YM-MA-2 was chosen for 3D bioprinting. Our data demonstrated that hydrogel products with designed shape and living chondrocytes have been printed by applying YM-MA-2 as the bioink carrying chondrocytes. After the YM-MA-2 hydrogel with encapsulated chondrocytes was implanted subcutaneously in nude mice for 2 weeks, GAG and COLII secretion was confirmed by histological staining in YM-MA-2-H, indicating that the YM derived bioink can be potentially applied to tissue engineering by employing a 3D printer of stereolithography.
Collapse
Affiliation(s)
- Yuting Huang
- College of Material Science and Engineering, Hunan University, Changsha 410082, People's Republic of China
| | - Zheng Zhou
- College of Biology, Hunan University, Changsha 410082, People's Republic of China
| | - Yingbing Hu
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, People's Republic of China
| | - Ning He
- State Key Laboratory of Advanced Design and Manufacture for Vehicle Body, Hunan University, Changsha 410082, People's Republic of China
| | - Jing Li
- State Key Laboratory of Advanced Design and Manufacture for Vehicle Body, Hunan University, Changsha 410082, People's Republic of China
| | - Xiaoxiao Han
- State Key Laboratory of Advanced Design and Manufacture for Vehicle Body, Hunan University, Changsha 410082, People's Republic of China
| | - Guoqun Zhao
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, People's Republic of China
| | - Hairong Liu
- College of Material Science and Engineering, Hunan University, Changsha 410082, People's Republic of China
| |
Collapse
|
137
|
Polonchuk L, Surija L, Lee MH, Sharma P, Liu Chung Ming C, Richter F, Ben-Sefer E, Rad MA, Mahmodi Sheikh Sarmast H, Shamery WA, Tran HA, Vettori L, Haeusermann F, Filipe EC, Rnjak-Kovacina J, Cox T, Tipper J, Kabakova I, Gentile C. Towards engineering heart tissues from bioprinted cardiac spheroids. Biofabrication 2021; 13. [PMID: 34265755 DOI: 10.1088/1758-5090/ac14ca] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/15/2021] [Indexed: 02/06/2023]
Abstract
Currentin vivoandin vitromodels fail to accurately recapitulate the human heart microenvironment for biomedical applications. This study explores the use of cardiac spheroids (CSs) to biofabricate advancedin vitromodels of the human heart. CSs were created from human cardiac myocytes, fibroblasts and endothelial cells (ECs), mixed within optimal alginate/gelatin hydrogels and then bioprinted on a microelectrode plate for drug testing. Bioprinted CSs maintained their structure and viability for at least 30 d after printing. Vascular endothelial growth factor (VEGF) promoted EC branching from CSs within hydrogels. Alginate/gelatin-based hydrogels enabled spheroids fusion, which was further facilitated by addition of VEGF. Bioprinted CSs contracted spontaneously and under stimulation, allowing to record contractile and electrical signals on the microelectrode plates for industrial applications. Taken together, our findings indicate that bioprinted CSs can be used to biofabricate human heart tissues for long termin vitrotesting. This has the potential to be used to study biochemical, physiological and pharmacological features of human heart tissue.
Collapse
Affiliation(s)
- Liudmila Polonchuk
- F Hoffmann-La Roche AG Research and Development Division, Pharmaceutical Sciences, Roche Innovation Center Basel, Grenzacherstrasse 124, Basel, Basel-Stadt CH-4070, Switzerland
| | - Lydia Surija
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia
| | - Min Ho Lee
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia
| | - Poonam Sharma
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia.,The University of Newcastle Faculty of Health and Medicine, University Drive, Callaghan, NSW 2308, Australia.,University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Clara Liu Chung Ming
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Florian Richter
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia
| | - Eitan Ben-Sefer
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Maryam Alsadat Rad
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Hadi Mahmodi Sheikh Sarmast
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Wafa Al Shamery
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Hien A Tran
- School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Laura Vettori
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Fabian Haeusermann
- F Hoffmann-La Roche AG Research and Development Division, Pharmaceutical Sciences, Roche Innovation Center Basel, Grenzacherstrasse 124, Basel, Basel-Stadt CH-4070, Switzerland
| | - Elysse C Filipe
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia.,St Vincent Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | | | - Thomas Cox
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia.,St Vincent Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Joanne Tipper
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Irina Kabakova
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Carmine Gentile
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia.,University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| |
Collapse
|
138
|
Tavafoghi M, Darabi MA, Mahmoodi M, Tutar R, Xu C, Mirjafari A, Billi F, Swieszkowski W, Nasrollahi F, Ahadian S, Hosseini V, Khademhosseini A, Ashammakhi N. Multimaterial bioprinting and combination of processing techniques towards the fabrication of biomimetic tissues and organs. Biofabrication 2021; 13. [PMID: 34130266 DOI: 10.1088/1758-5090/ac0b9a] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022]
Abstract
Tissue reconstruction requires the utilization of multiple biomaterials and cell types to replicate the delicate and complex structure of native tissues. Various three-dimensional (3D) bioprinting techniques have been developed to fabricate customized tissue structures; however, there are still significant challenges, such as vascularization, mechanical stability of printed constructs, and fabrication of gradient structures to be addressed for the creation of biomimetic and complex tissue constructs. One approach to address these challenges is to develop multimaterial 3D bioprinting techniques that can integrate various types of biomaterials and bioprinting capabilities towards the fabrication of more complex structures. Notable examples include multi-nozzle, coaxial, and microfluidics-assisted multimaterial 3D bioprinting techniques. More advanced multimaterial 3D printing techniques are emerging, and new areas in this niche technology are rapidly evolving. In this review, we briefly introduce the basics of individual 3D bioprinting techniques and then discuss the multimaterial 3D printing techniques that can be developed based on combination of these techniques for the engineering of complex and biomimetic tissue constructs. We also discuss the perspectives and future directions to develop state-of-the-art multimaterial 3D bioprinting techniques for engineering tissues and organs.
Collapse
Affiliation(s)
- Maryam Tavafoghi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Mahboobeh Mahmoodi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Rumeysa Tutar
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa Avcılar, Istanbul 34320, Turkey
| | - Chun Xu
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,School of Dentistry, The University of Queensland, Brisbane, Australia
| | - Arshia Mirjafari
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America
| | - Fabrizio Billi
- UCLA/OIC Department of Orthopaedic Surgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States of America
| | - Wojciech Swieszkowski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Fatemeh Nasrollahi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Vahid Hosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America.,Department of Chemical Engineering, University of California, Los Angeles, CA, United States of America
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America.,Department of Biomedical Engineering, College of Engineering, Michigan State University, MI, United States of America
| |
Collapse
|
139
|
Pozzi S, Scomparin A, Israeli Dangoor S, Rodriguez Ajamil D, Ofek P, Neufeld L, Krivitsky A, Vaskovich-Koubi D, Kleiner R, Dey P, Koshrovski-Michael S, Reisman N, Satchi-Fainaro R. Meet me halfway: Are in vitro 3D cancer models on the way to replace in vivo models for nanomedicine development? Adv Drug Deliv Rev 2021; 175:113760. [PMID: 33838208 DOI: 10.1016/j.addr.2021.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
Abstract
The complexity and diversity of the biochemical processes that occur during tumorigenesis and metastasis are frequently over-simplified in the traditional in vitro cell cultures. Two-dimensional cultures limit researchers' experimental observations and frequently give rise to misleading and contradictory results. Therefore, in order to overcome the limitations of in vitro studies and bridge the translational gap to in vivo applications, 3D models of cancer were developed in the last decades. The three dimensions of the tumor, including its cellular and extracellular microenvironment, are recreated by combining co-cultures of cancer and stromal cells in 3D hydrogel-based growth factors-inclusive scaffolds. More complex 3D cultures, containing functional blood vasculature, can integrate in the system external stimuli (e.g. oxygen and nutrient deprivation, cytokines, growth factors) along with drugs, or other therapeutic compounds. In this scenario, cell signaling pathways, metastatic cascade steps, cell differentiation and self-renewal, tumor-microenvironment interactions, and precision and personalized medicine, are among the wide range of biological applications that can be studied. Here, we discuss a broad variety of strategies exploited by scientists to create in vitro 3D cancer models that resemble as much as possible the biology and patho-physiology of in vivo tumors and predict faithfully the treatment outcome.
Collapse
Affiliation(s)
- Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy
| | - Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniel Rodriguez Ajamil
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lena Neufeld
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniella Vaskovich-Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noa Reisman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
140
|
Shao L, Hou R, Zhu Y, Yao Y. Pre-shear bioprinting of highly oriented porous hydrogel microfibers to construct anisotropic tissues. Biomater Sci 2021; 9:6763-6771. [PMID: 34286720 DOI: 10.1039/d1bm00695a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Anisotropic tissues in vivo have special structural characteristics and biological functions. Nowadays, bioprinting is widely used in tissue engineering and an effective way to process cell-laden hydrogels. However, the direct bioprinting of oriented cell-laden hydrogel structures to engineer anisotropic tissues is still difficult. Meanwhile, the inherent dense micropore network after the gelation of hydrogel-based bioinks usually limits the normal growth of encapsulated cells due to the inadequate supply of nutrient/oxygen. Herein, we proposed a pre-shear bioprinting strategy of highly oriented porous hydrogel microfibers to construct anisotropic tissues. Firstly, based on the phase separation of viscous high-molecular compound mixtures, we utilized a general viscous porous bioink paradigm, e.g., mixing a polymer thickener (PEO) with a hydrogel precursor (GelMA) with excellent biological properties. Secondly, based on the shear-oriented property of the viscous porous bioink, we designed the pre-shear in situ coaxial bioprinting of highly oriented porous hydrogel microfibers. The viscous porous bioink (GelMA/PEO) was shear-oriented through an injection tube and pumped into the inner needle of a coaxial nozzle. When GelMA/PEO passed through a transparent glass tube connected to the coaxial nozzle, GelMA can be in situ photo-crosslinked to form highly oriented porous microfibers. In addition, we showed the manufacturing of heterogeneous oriented microfibers and the manual assembly of microfibers, and within oriented microfibers, different cells or co-cultured cells exhibited highly oriented growth behaviors similar to that in vivo. As far as we know, the direct bioprinting of anisotropic tissues through high orientation induced by pre-shearing is firstly reported in our study. We believe that the pre-shear bioprinting strategy of anisotropic tissues will open more avenues for further biomedical research.
Collapse
Affiliation(s)
- Lei Shao
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo, 315211, China. and State Key Laboratory of Fluid Power and Mechatronic Systems, College of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ruixia Hou
- School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Yabin Zhu
- School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Yudong Yao
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
141
|
Kilian D, Sembdner P, Bretschneider H, Ahlfeld T, Mika L, Lützner J, Holtzhausen S, Lode A, Stelzer R, Gelinsky M. 3D printing of patient-specific implants for osteochondral defects: workflow for an MRI-guided zonal design. Biodes Manuf 2021. [DOI: 10.1007/s42242-021-00153-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Abstract
Magnetic resonance imaging (MRI) is a common clinical practice to visualize defects and to distinguish different tissue types and pathologies in the human body. So far, MRI data have not been used to model and generate a patient-specific design of multilayered tissue substitutes in the case of interfacial defects. For orthopedic cases that require highly individual surgical treatment, implant fabrication by additive manufacturing holds great potential. Extrusion-based techniques like 3D plotting allow the spatially defined application of several materials, as well as implementation of bioprinting strategies. With the example of a typical multi-zonal osteochondral defect in an osteochondritis dissecans (OCD) patient, this study aimed to close the technological gap between MRI analysis and the additive manufacturing process of an implant based on different biomaterial inks. A workflow was developed which covers the processing steps of MRI-based defect identification, segmentation, modeling, implant design adjustment, and implant generation. A model implant was fabricated based on two biomaterial inks with clinically relevant properties that would allow for bioprinting, the direct embedding of a patient’s own cells in the printing process. As demonstrated by the geometric compatibility of the designed and fabricated model implant in a stereolithography (SLA) model of lesioned femoral condyles, a novel versatile CAD/CAM workflow was successfully established that opens up new perspectives for the treatment of multi-zonal (osteochondral) defects.
Graphic abstract
Collapse
|
142
|
Evaluation of 3D-Printing Scaffold Fabrication on Biosynthetic Medium-Chain-Length Polyhydroxyalkanoate Terpolyester as Biomaterial-Ink. Polymers (Basel) 2021; 13:polym13142222. [PMID: 34300981 PMCID: PMC8309464 DOI: 10.3390/polym13142222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 11/17/2022] Open
Abstract
Currently, the selection of materials for tissue engineering scaffolds is still limited because some tissues require flexible and compatible materials with human cells. Medium-chain-length polyhydroxyalkanoate (MCL-PHA) synthesized in microorganisms is an interesting polymer for use in this area and has elastomeric properties compatible with the human body. MCL-PHAs are elastomers with biodegradability and cellular compatibility, making them an attractive material for fabricating soft tissue that requires high elasticity. In this research, MCL-PHA was produced by fed-batch fermentation that Pseudomonas Putida ATCC 47054 was cultured to accumulate MCL-PHA by using glycerol and sodium octanoate as carbon sources. The amounts of dry cell density, MCL-PHA product per dry cells, and MCL-PHA productivity were at 15 g/L, 27%, and 0.067 g/L/h, respectively, and the components of MCL-PHA consisting of 3-hydroxydecanoate (3HD) 64.5%, 3-hydroxyoctanoate (3HO) 32.2%, and 3-hydroxyhexanoate (3HHx) 3.3%. The biosynthesized MCL-PHA terpolyester has a relatively low melting temperature, low crystallinity, and high ductility at 52 °C, 15.7%, and 218%, respectively, and considering as elastomeric polyester. The high-resolution scaffold of MCL-PHA terpolyester biomaterial-ink (approximately 0.36 mm porous size) could be printed in a selected condition with a 3D printer, similar to the optimum pore size for cell attachment and proliferation. The rheological characteristic of this MCL-PHA biomaterial-ink exhibits shear-thinning behavior, leading to good shape fidelity. The study results yielded a condition capable of fabricating an elastomer scaffold of the MCL-PHA terpolyester, giving rise to the ideal soft tissue engineering application.
Collapse
|
143
|
Zhu J, Wang Y, Zhong L, Pan F, Wang J. Advances in tissue engineering of vasculature through three-dimensional bioprinting. Dev Dyn 2021; 250:1717-1738. [PMID: 34115420 DOI: 10.1002/dvdy.385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/07/2021] [Accepted: 06/03/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND A significant challenge facing tissue engineering is the fabrication of vasculature constructs which contains vascularized tissue constructs to recapitulate viable, complex and functional organs or tissues, and free-standing vascular structures potentially providing clinical applications in the future. Three-dimensional (3D) bioprinting has emerged as a promising technology, possessing a number of merits that other conventional biofabrication methods do not have. Over the last decade, 3D bioprinting has contributed a variety of techniques and strategies to generate both vascularized tissue constructs and free-standing vascular structures. RESULTS This review focuses on different strategies to print two kinds of vasculature constructs, namely vascularized tissue constructs and vessel-like tubular structures, highlighting the feasibility and shortcoming of the current methods for vasculature constructs fabrication. Generally, both direct printing and indirect printing can be employed in vascularized tissue engineering. Direct printing allows for structural fabrication with synchronous cell seeding, while indirect printing is more effective in generating complex architecture. During the fabrication process, 3D bioprinting techniques including extrusion bioprinting, inkjet bioprinting and light-assisted bioprinting should be selectively implemented to exert advantages and obtain the desirable tissue structure. Also, appropriate cells and biomaterials matter a lot to match various bioprinting techniques and thus achieve successful fabrication of specific vasculature constructs. CONCLUSION The 3D bioprinting has been developed to help provide various fabrication techniques, devoting to producing structurally stable, physiologically relevant, and biologically appealing constructs. However, although the optimization of biomaterials and innovation of printing strategies may improve the fabricated vessel-like structures, 3D bioprinting is still in the infant period and has a great gap between in vitro trials and in vivo applications. The article reviews the present achievement of 3D bioprinting in generating vasculature constructs and also provides perspectives on future directions of advanced vasculature constructs fabrication.
Collapse
Affiliation(s)
- Junjin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuting Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linna Zhong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fangwei Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
144
|
Moghaddam AS, Khonakdar HA, Arjmand M, Jafari SH, Bagher Z, Moghaddam ZS, Chimerad M, Sisakht MM, Shojaei S. Review of Bioprinting in Regenerative Medicine: Naturally Derived Bioinks and Stem Cells. ACS APPLIED BIO MATERIALS 2021; 4:4049-4070. [PMID: 35006822 DOI: 10.1021/acsabm.1c00219] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerative medicine offers the potential to repair or substitute defective tissues by constructing active tissues to address the scarcity and demands for transplantation. The method of forming 3D constructs made up of biomaterials, cells, and biomolecules is called bioprinting. Bioprinting of stem cells provides the ability to reliably recreate tissues, organs, and microenvironments to be used in regenerative medicine. 3D bioprinting is a technique that uses several biomaterials and cells to tailor a structure with clinically relevant geometries and sizes. This technique's promise is demonstrated by 3D bioprinted tissues, including skin, bone, cartilage, and cardiovascular, corneal, hepatic, and adipose tissues. Several bioprinting methods have been combined with stem cells to effectively produce tissue models, including adult stem cells, embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and differentiation techniques. In this review, technological challenges of printed stem cells using prevalent naturally derived bioinks (e.g., carbohydrate polymers and protein-based polymers, peptides, and decellularized extracellular matrix), recent advancements, leading companies, and clinical trials in the field of 3D bioprinting are delineated.
Collapse
Affiliation(s)
- Abolfazl Salehi Moghaddam
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 11155-4593, Iran
| | - Hossein Ali Khonakdar
- Leibniz Institute of Polymer Research Dresden, Hohe Straße 6, Dresden D-01069, Germany.,Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
| | - Mohammad Arjmand
- Nanomaterials and Polymer Nanocomposites Laboratory, School of Engineering, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - Seyed Hassan Jafari
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 11155-4593, Iran
| | - Zohreh Bagher
- ENT and Head & Neck Research Centre and Department, The Five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Zahra Salehi Moghaddam
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Mohammadreza Chimerad
- School of Mechanical Engineering, Iran University of Science and Technology, Tehran 16844, Iran
| | - Mahsa Mollapour Sisakht
- Stem Cell and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran 19379-57511, Iran.,Department of Biochemistry, Erasmus University Medical Center, Rotterdam 3000 DR, The Netherlands
| | - Shahrokh Shojaei
- Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, PO Box 13185/768, Tehran 15689-37813, Iran.,Stem Cells Research Center, Tissue Engineering and Regenerative Medicine Institute, Islamic Azad University, Central Tehran Branch, PO Box 13185-768, Tehran 15689-37813, Iran
| |
Collapse
|
145
|
Fabrication of Microfluidic Devices for Emulsion Formation by Microstereolithography. Molecules 2021; 26:molecules26092817. [PMID: 34068649 PMCID: PMC8126101 DOI: 10.3390/molecules26092817] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/10/2021] [Accepted: 04/27/2021] [Indexed: 01/22/2023] Open
Abstract
Droplet microfluidics—the art and science of forming droplets—has been revolutionary for high-throughput screening, directed evolution, single-cell sequencing, and material design. However, traditional fabrication techniques for microfluidic devices suffer from several disadvantages, including multistep processing, expensive facilities, and limited three-dimensional (3D) design flexibility. High-resolution additive manufacturing—and in particular, projection micro-stereolithography (PµSL)—provides a promising path for overcoming these drawbacks. Similar to polydimethylsiloxane-based microfluidics 20 years ago, 3D printing methods, such as PµSL, have provided a path toward a new era of microfluidic device design. PµSL greatly simplifies the device fabrication process, especially the access to truly 3D geometries, is cost-effective, and it enables multimaterial processing. In this review, we discuss both the basics and recent innovations in PµSL; the material basis with emphasis on custom-made photopolymer formulations; multimaterial 3D printing; and, 3D-printed microfluidic devices for emulsion formation as our focus application. Our goal is to support researchers in setting up their own PµSL system to fabricate tailor-made microfluidics.
Collapse
|
146
|
Kirillova A, Yeazel TR, Asheghali D, Petersen SR, Dort S, Gall K, Becker ML. Fabrication of Biomedical Scaffolds Using Biodegradable Polymers. Chem Rev 2021; 121:11238-11304. [PMID: 33856196 DOI: 10.1021/acs.chemrev.0c01200] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Degradable polymers are used widely in tissue engineering and regenerative medicine. Maturing capabilities in additive manufacturing coupled with advances in orthogonal chemical functionalization methodologies have enabled a rapid evolution of defect-specific form factors and strategies for designing and creating bioactive scaffolds. However, these defect-specific scaffolds, especially when utilizing degradable polymers as the base material, present processing challenges that are distinct and unique from other classes of materials. The goal of this review is to provide a guide for the fabrication of biodegradable polymer-based scaffolds that includes the complete pathway starting from selecting materials, choosing the correct fabrication method, and considering the requirements for tissue specific applications of the scaffold.
Collapse
Affiliation(s)
- Alina Kirillova
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Taylor R Yeazel
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Darya Asheghali
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Shannon R Petersen
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Sophia Dort
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Ken Gall
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Matthew L Becker
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States.,Department of Chemistry, Duke University, Durham, North Carolina 27708, United States.,Departments of Biomedical Engineering and Orthopaedic Surgery, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
147
|
Taymour R, Kilian D, Ahlfeld T, Gelinsky M, Lode A. 3D bioprinting of hepatocytes: core-shell structured co-cultures with fibroblasts for enhanced functionality. Sci Rep 2021; 11:5130. [PMID: 33664366 PMCID: PMC7933206 DOI: 10.1038/s41598-021-84384-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/16/2021] [Indexed: 01/31/2023] Open
Abstract
With the aim of understanding and recapitulating cellular interactions of hepatocytes in their physiological microenvironment and to generate an artificial 3D in vitro model, a co-culture system using 3D extrusion bioprinting was developed. A bioink based on alginate and methylcellulose (algMC) was first shown to be suitable for bioprinting of hepatocytes; the addition of Matrigel to algMC enhanced proliferation and morphology of them in monophasic scaffolds. Towards a more complex system that allows studying cellular interactions, we applied core-shell bioprinting to establish tailored 3D co-culture models for hepatocytes. The bioinks were specifically functionalized with natural matrix components (based on human plasma, fibrin or Matrigel) and used to co-print fibroblasts and hepatocytes in a spatially defined, coaxial manner. Fibroblasts acted as supportive cells for co-cultured hepatocytes, stimulating the expression of certain biomarkers of hepatocytes like albumin. Furthermore, matrix functionalization positively influenced both cell types in their respective compartments by enhancing their adhesion, viability, proliferation and function. In conclusion, we established a functional co-culture model with independently tunable compartments for different cell types via core-shell bioprinting. This provides the basis for more complex in vitro models allowing co-cultivation of hepatocytes with other liver-specific cell types to closely resemble the liver microenvironment.
Collapse
Affiliation(s)
- Rania Taymour
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - David Kilian
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Tilman Ahlfeld
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
| |
Collapse
|
148
|
Shiwarski DJ, Hudson AR, Tashman JW, Feinberg AW. Emergence of FRESH 3D printing as a platform for advanced tissue biofabrication. APL Bioeng 2021; 5:010904. [PMID: 33644626 PMCID: PMC7889293 DOI: 10.1063/5.0032777] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022] Open
Abstract
In tissue engineering, an unresolved challenge is how to build complex 3D scaffolds in order to recreate the structure and function of human tissues and organs. Additive manufacturing techniques, such as 3D bioprinting, have the potential to build biological material with unprecedented spatial control; however, printing soft biological materials in air often results in poor fidelity. Freeform Reversible Embedding of Suspended Hydrogels (FRESH) is an embedded printing approach that solves this problem by extruding bioinks within a yield-stress support bath that holds the bioinks in place until cured. In this Perspective, we discuss the challenges of 3D printing soft and liquid-like bioinks and the emergence for FRESH and related embedded printing techniques as a solution. This includes the development of FRESH and embedded 3D printing within the bioprinting field and the rapid growth in adoption, as well as the advantages of FRESH printing for biofabrication and the new research results this has enabled. Specific focus is on the customizability of the FRESH printing technique where the chemical composition of the yield-stress support bath and aqueous phase crosslinker can all be tailored for printing a wide range of bioinks in complex 3D structures. Finally, we look ahead at the future of FRESH printing, discussing both the challenges and the opportunities that we see as the biofabrication field develops.
Collapse
Affiliation(s)
- Daniel J. Shiwarski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| | - Andrew R. Hudson
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| | - Joshua W. Tashman
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| | | |
Collapse
|
149
|
Cooke ME, Rosenzweig DH. The rheology of direct and suspended extrusion bioprinting. APL Bioeng 2021; 5:011502. [PMID: 33564740 PMCID: PMC7864677 DOI: 10.1063/5.0031475] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/18/2021] [Indexed: 12/11/2022] Open
Abstract
Bioprinting is a tool increasingly used in tissue engineering laboratories around the world. As an extension to classic tissue engineering, it enables high levels of control over the spatial deposition of cells, materials, and other factors. It is a field with huge promise for the production of implantable tissues and even organs, but the availability of functional bioinks is a barrier to success. Extrusion bioprinting is the most commonly used technique, where high-viscosity solutions of materials and cells are required to ensure good shape fidelity of the printed tissue construct. This is contradictory to hydrogels used in tissue engineering, which are generally of low viscosity prior to cross-linking to ensure cell viability, making them not directly translatable to bioprinting. This review provides an overview of the important rheological parameters for bioinks and methods to assess printability, as well as the effect of bioink rheology on cell viability. Developments over the last five years in bioink formulations and the use of suspended printing to overcome rheological limitations are then discussed.
Collapse
|
150
|
Aldana AA, Houben S, Moroni L, Baker MB, Pitet LM. Trends in Double Networks as Bioprintable and Injectable Hydrogel Scaffolds for Tissue Regeneration. ACS Biomater Sci Eng 2021; 7:4077-4101. [DOI: 10.1021/acsbiomaterials.0c01749] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Ana A. Aldana
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Sofie Houben
- Advanced Functional Polymers Group, Department of Chemistry, Institute for Materials Research (IMO), Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Matthew B. Baker
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Louis M. Pitet
- Advanced Functional Polymers Group, Department of Chemistry, Institute for Materials Research (IMO), Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium
| |
Collapse
|