1
|
Tanadchangsaeng N, Pasanaphong K, Tawonsawatruk T, Rattanapinyopituk K, Tangketsarawan B, Rawiwet V, Kongchanagul A, Srikaew N, Yoyruerop T, Panupinthu N, Sangpayap R, Panaksri A, Boonyagul S, Hemstapat R. 3D bioprinting of fish skin-based gelatin methacryloyl (GelMA) bio-ink for use as a potential skin substitute. Sci Rep 2024; 14:23240. [PMID: 39369014 PMCID: PMC11455937 DOI: 10.1038/s41598-024-73774-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Gelatin methacryloyl (GelMA), typically derived from mammalian sources, has recently emerged as an ideal bio-ink for three-dimensional (3D) bioprinting. Herein, we developed a fish skin-based GelMA bio-ink for the fabrication of a 3D GelMA skin substitute with a 3D bioprinter. Several concentrations of methacrylic acid anhydride were used to fabricate GelMA, in which their physical-mechanical properties were assessed. This fish skin-based GelMA bio-ink was loaded with human adipose tissue-derived mesenchymal stromal cells (ASCs) and human platelet lysate (HPL) and then printed to obtain 3D ASCs + HPL-loaded GelMA scaffolds. Cell viability test and a preliminary investigation of its effectiveness in promoting wound closure were evaluated in a critical-sized full thickness skin defect in a rat model. The cell viability results showed that the number of ASCs increased significantly within the 3D GelMA hydrogel scaffold, indicating its biocompatibility property. In vivo results demonstrated that ASCs + HPL-loaded GelMA scaffolds could delay wound contraction, markedly enhanced collagen deposition, and promoted the formation of new blood vessels, especially at the wound edge, compared to the untreated group. Therefore, this newly fish skin-based GelMA bio-ink developed in this study has the potential to be utilized for the printing of 3D GelMA skin substitutes.
Collapse
Affiliation(s)
| | | | - Tulyapruek Tawonsawatruk
- Department of Orthopaedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kasem Rattanapinyopituk
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | | | - Visut Rawiwet
- Central Animal Facility, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Alita Kongchanagul
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Bangkok, Thailand
| | - Narongrit Srikaew
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thanaporn Yoyruerop
- Mahidol University-Frontier Research Facility (MU-FRF), Mahidol University, Nakhon Pathom, Thailand
| | - Nattapon Panupinthu
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ratirat Sangpayap
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Anuchan Panaksri
- College of Biomedical Engineering, Rangsit University, Pathum Thani, Thailand
| | - Sani Boonyagul
- College of Biomedical Engineering, Rangsit University, Pathum Thani, Thailand
| | - Ruedee Hemstapat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
2
|
Meijer E, Giles R, van Dijk CGM, Maringanti R, Wissing TB, Appels Y, Chrifi I, Crielaard H, Verhaar MC, Smits AI, Cheng C. Effect of Mechanical Stimuli on the Phenotypic Plasticity of Induced Pluripotent Stem-Cell-Derived Vascular Smooth Muscle Cells in a 3D Hydrogel. ACS APPLIED BIO MATERIALS 2023; 6:5716-5729. [PMID: 38032545 PMCID: PMC10731661 DOI: 10.1021/acsabm.3c00840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 12/01/2023]
Abstract
Introduction: Vascular smooth muscle cells (VSMCs) play a pivotal role in vascular homeostasis, with dysregulation leading to vascular complications. Human-induced pluripotent stem-cell (hiPSC)-derived VSMCs offer prospects for personalized disease modeling and regenerative strategies. Current research lacks comparative studies on the impact of three-dimensional (3D) substrate properties under cyclic strain on phenotypic adaptation in hiPSC-derived VSMCs. Here, we aim to investigate the impact of intrinsic substrate properties, such as the hydrogel's elastic modulus and cross-linking density in a 3D static and dynamic environment, on the phenotypical adaptation of human mural cells derived from hiPSC-derived organoids (ODMCs), compared to aortic VSMCs. Methods and results: ODMCs were cultured in two-dimensional (2D) conditions with synthetic or contractile differentiation medium or in 3D Gelatin Methacryloyl (GelMa) substrates with varying degrees of functionalization and percentages to modulate Young's modulus and cross-linking density. Cells in 3D substrates were exposed to cyclic, unidirectional strain. Phenotype characterization was conducted using specific markers through immunofluorescence and gene expression analysis. Under static 2D culture, ODMCs derived from hiPSCs exhibited a VSMC phenotype, expressing key mural markers, and demonstrated a level of phenotypic plasticity similar to primary human VSMCs. In static 3D culture, a substrate with a higher Young's modulus and cross-linking density promoted a contractile phenotype in ODMCs and VSMCs. Dynamic stimulation in the 3D substrate promoted a switch toward a contractile phenotype in both cell types. Conclusion: Our study demonstrates phenotypic plasticity of human ODMCs in response to 2D biological and 3D mechanical stimuli that equals that of primary human VSMCs. These findings may contribute to the advancement of tailored approaches for vascular disease modeling and regenerative strategies.
Collapse
Affiliation(s)
- Elana
M. Meijer
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Rachel Giles
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Christian G. M. van Dijk
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Ranganath Maringanti
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
- Experimental
Cardiology, Department of Cardiology, Thorax
Center Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Tamar B. Wissing
- Department
of Biomedical Engineering, Eindhoven University
of Technology; Eindhoven 5612 AE, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology; Eindhoven 5612 AE, The Netherlands
| | - Ymke Appels
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Ihsan Chrifi
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
- Experimental
Cardiology, Department of Cardiology, Thorax
Center Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Hanneke Crielaard
- Department
of Biomedical Engineering, Erasmus Medical
Center, Rotterdam 3000 CA, The Netherlands
| | - Marianne C. Verhaar
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Anthal I.P.M. Smits
- Department
of Biomedical Engineering, Eindhoven University
of Technology; Eindhoven 5612 AE, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology; Eindhoven 5612 AE, The Netherlands
| | - Caroline Cheng
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
- Experimental
Cardiology, Department of Cardiology, Thorax
Center Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| |
Collapse
|
3
|
Schwab A, Wesdorp MA, Xu J, Abinzano F, Loebel C, Falandt M, Levato R, Eglin D, Narcisi R, Stoddart MJ, Malda J, Burdick JA, D'Este M, van Osch GJ. Modulating design parameters to drive cell invasion into hydrogels for osteochondral tissue formation. J Orthop Translat 2023; 41:42-53. [PMID: 37691639 PMCID: PMC10485598 DOI: 10.1016/j.jot.2023.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/08/2023] [Accepted: 07/03/2023] [Indexed: 09/12/2023] Open
Abstract
Background The use of acellular hydrogels to repair osteochondral defects requires cells to first invade the biomaterial and then to deposit extracellular matrix for tissue regeneration. Due to the diverse physicochemical properties of engineered hydrogels, the specific properties that allow or even improve the behaviour of cells are not yet clear. The aim of this study was to investigate the influence of various physicochemical properties of hydrogels on cell migration and related tissue formation using in vitro, ex vivo and in vivo models. Methods Three hydrogel platforms were used in the study: Gelatine methacryloyl (GelMA) (5% wt), norbornene hyaluronic acid (norHA) (2% wt) and tyramine functionalised hyaluronic acid (THA) (2.5% wt). GelMA was modified to vary the degree of functionalisation (DoF 50% and 80%), norHA was used with varied degradability via a matrix metalloproteinase (MMP) degradable crosslinker and THA was used with the addition of collagen fibrils. The migration of human mesenchymal stromal cells (hMSC) in hydrogels was studied in vitro using a 3D spheroid migration assay over 48h. In addition, chondrocyte migration within and around hydrogels was investigated in an ex vivo bovine cartilage ring model (three weeks). Finally, tissue repair within osteochondral defects was studied in a semi-orthotopic in vivo mouse model (six weeks). Results A lower DoF of GelMA did not affect cell migration in vitro (p = 0.390) and led to a higher migration score ex vivo (p < 0.001). The introduction of a MMP degradable crosslinker in norHA hydrogels did not improve cell infiltration in vitro or in vivo. The addition of collagen to THA resulted in greater hMSC migration in vitro (p = 0.031) and ex vivo (p < 0.001). Hydrogels that exhibited more cell migration in vitro or ex vivo also showed more tissue formation in the osteochondral defects in vivo, except for the norHA group. Whereas norHA with a degradable crosslinker did not improve cell migration in vitro or ex vivo, it did significantly increase tissue formation in vivo compared to the non-degradable crosslinker (p < 0.001). Conclusion The modification of hydrogels by adapting DoF, use of a degradable crosslinker or including fibrillar collagen can control and improve cell migration and tissue formation for osteochondral defect repair. This study also emphasizes the importance of performing both in vitro and in vivo testing of biomaterials, as, depending on the material, the results might be affected by the model used.The translational potential of this article: This article highlights the potential of using acellular hydrogels to repair osteochondral defects, which are common injuries in orthopaedics. The study provides a deeper understanding of how to modify the properties of hydrogels to control cell migration and tissue formation for osteochondral defect repair. The results of this article also highlight that the choice of the used laboratory model can affect the outcome. Testing hydrogels in different models is thus advised for successful translation of laboratory results to the clinical application.
Collapse
Affiliation(s)
- Andrea Schwab
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
| | - Marinus A. Wesdorp
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Jietao Xu
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Florencia Abinzano
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc Falandt
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, the Netherlands
| | - Riccardo Levato
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, the Netherlands
| | - David Eglin
- Mines Saint-Etienne, University Jean Monnet, INSERM, UMR 1059, Saint-Etienne, France
- Advanced Organ Bioengineering and Therapeutics, Faculty of Science and Technology, TechMed Center, University of Twente, Enschede, the Netherlands
| | - Roberto Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | | | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Matteo D'Este
- AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Delft, the Netherlands
| |
Collapse
|
4
|
A vertical additive-lathe printing system for the fabrication of tubular constructs using gelatin methacryloyl hydrogel. J Mech Behav Biomed Mater 2023; 139:105665. [PMID: 36640542 DOI: 10.1016/j.jmbbm.2023.105665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/26/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
Reproducing both the mechanical and biological performance of native blood vessels remains an ongoing challenge in vascular tissue engineering. Additive-lathe printing offers an attractive method of fabricating long tubular constructs as a potential vascular graft for the treatment of cardiovascular diseases. Printing hydrogels onto rotating horizontal mandrels often leads to sagging, resulting in poor and variable mechanical properties. In this study, an additive-lathe printing system with a vertical mandrel to fabricate tubular constructs is presented. Various concentrations of gelatin methacryloyl (gelMA) hydrogel were used to print grafts on the rotating mandrel in a helical pattern. The printing parameters were selected to achieve the bonding of consecutive gelMA filaments to improve the quality of the printed graft. The hydrogel filaments were fused properly under the action of gravity on the vertical mandrel. Thus, the vertical additive-lathe printing system was used to print uniform wall thickness grafts, eliminating the hydrogel sagging problem. Tensile testing performed in both circumferential and longitudinal direction revealed that the anisotropic properties of printed gelMA constructs were similar to those observed in the native blood vessels. In addition, no leakage was detected through the walls of the gelMA grafts during burst pressure measurement. Therefore, the current printing setup could be utilized to print vascular grafts for the treatment of cardiovascular diseases.
Collapse
|
5
|
Schulze F, Lang A, Schoon J, Wassilew GI, Reichert J. Scaffold Guided Bone Regeneration for the Treatment of Large Segmental Defects in Long Bones. Biomedicines 2023; 11:biomedicines11020325. [PMID: 36830862 PMCID: PMC9953456 DOI: 10.3390/biomedicines11020325] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Bone generally displays a high intrinsic capacity to regenerate. Nonetheless, large osseous defects sometimes fail to heal. The treatment of such large segmental defects still represents a considerable clinical challenge. The regeneration of large bone defects often proves difficult, since it relies on the formation of large amounts of bone within an environment impedimental to osteogenesis, characterized by soft tissue damage and hampered vascularization. Consequently, research efforts have concentrated on tissue engineering and regenerative medical strategies to resolve this multifaceted challenge. In this review, we summarize, critically evaluate, and discuss present approaches in light of their clinical relevance; we also present future advanced techniques for bone tissue engineering, outlining the steps to realize for their translation from bench to bedside. The discussion includes the physiology of bone healing, requirements and properties of natural and synthetic biomaterials for bone reconstruction, their use in conjunction with cellular components and suitable growth factors, and strategies to improve vascularization and the translation of these regenerative concepts to in vivo applications. We conclude that the ideal all-purpose material for scaffold-guided bone regeneration is currently not available. It seems that a variety of different solutions will be employed, according to the clinical treatment necessary.
Collapse
Affiliation(s)
- Frank Schulze
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Annemarie Lang
- Departments of Orthopaedic Surgery & Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Janosch Schoon
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Georgi I. Wassilew
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Johannes Reichert
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
- Correspondence: ; Tel.: +49-3834-86-22530
| |
Collapse
|
6
|
de Silva L, Bernal PN, Rosenberg A, Malda J, Levato R, Gawlitta D. Biofabricating the vascular tree in engineered bone tissue. Acta Biomater 2023; 156:250-268. [PMID: 36041651 DOI: 10.1016/j.actbio.2022.08.051] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2023]
Abstract
The development of tissue engineering strategies for treatment of large bone defects has become increasingly relevant, given the growing demand for bone substitutes. Native bone is composed of a dense vascular network necessary for the regulation of bone development, regeneration and homeostasis. A major obstacle in fabricating living, clinically relevant-sized bone mimics (1-10 cm3) is the limited supply of nutrients, including oxygen to the core of the construct. Therefore, strategies to support vascularization are pivotal for the development of tissue engineered bone constructs. Creating a functional bone construct integrated with a vascular network, capable of delivering the necessary nutrients for optimal tissue development is imperative for translation into the clinics. The vascular system is composed of a complex network that runs throughout the body in a tree-like hierarchical branching fashion. A significant challenge for tissue engineering approaches lies in mimicking the intricate, multi-scale structures consisting of larger vessels (macro-vessels) which interconnect with multiple sprouting vessels (microvessels) in a closed network. The advent of biofabrication has enabled complex, out of plane channels to be generated and has laid the groundwork for the creation of multi-scale vasculature in recent years. This review highlights the key state-of-the-art achievements for the development of vascular networks of varying scales in the field of biofabrication with a particular focus for its application in developing a functional tissue engineered bone construct. STATEMENT OF SIGNIFICANCE: There is a growing need for bone substitutes to overcome the limited supply of patient-derived bone. Bone tissue engineering aims to overcome this by combining stem cells with scaffolds to restore missing bone. The current bottleneck in upscaling is the lack of an integrated vascular network, required for the delivery of nutrients to cells. 3D bioprinting techniques has enabled the creation of complex hollow structures of varying dimensions that resemble native blood vessels. The convergence of multiple materials, cell types and fabrication approaches, opens the possibility of developing clinically-relevant sized vascularized bone constructs. This review provides an up-to-date insight of the technologies currently available for the generation of complex vascular networks, with a focus on their application in bone tissue engineering.
Collapse
Affiliation(s)
- Leanne de Silva
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands.
| | - Paulina N Bernal
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands
| | - Ajw Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands
| | - Jos Malda
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, the Netherlands
| | - Riccardo Levato
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, the Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands
| |
Collapse
|
7
|
Gehlen J, Qiu W, Schädli GN, Müller R, Qin XH. Tomographic volumetric bioprinting of heterocellular bone-like tissues in seconds. Acta Biomater 2023; 156:49-60. [PMID: 35718102 DOI: 10.1016/j.actbio.2022.06.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/24/2022] [Accepted: 06/09/2022] [Indexed: 01/18/2023]
Abstract
Tomographic volumetric bioprinting (VBP) has recently emerged as a powerful tool for rapid solidification of cell-laden hydrogel constructs within seconds. However, its practical applications in tissue engineering requires a detailed understanding of how different printing parameters (concentration of resins, laser dose) affect cell activity and tissue formation. Herein, we explore a new application of VBP in bone tissue engineering by merging a soft gelatin methacryloyl (GelMA) bioresin (<5 kPa) with 3D endothelial co-culture to generate heterocellular bone-like constructs with enhanced functionality. To this, a series of bioresins with varying concentrations of GelMA and lithium Phenyl(2,4,6-trimethylbenzoyl)phosphinate (LAP) photoinitiator were formulated and characterized in terms of photo-reactivity, printability and cell-compatibility. A bioresin with 5% GelMA and 0.05% LAP was identified as the optimal formulation for VBP of complex perfusable constructs within 30 s at high cell viability (>90%). The fidelity was validated by micro-computed tomography and confocal microscopy. Compared to 10% GelMA, this bioresin provided a softer and more permissive environment for osteogenic differentiation of human mesenchymal stem cells (hMSCs). The expression of osteoblastic markers (collagen-I, ALP, osteocalcin) and osteocytic markers (podoplanin, Dmp1) was monitored for 42 days. After 21 days, early osteocytic markers were significantly increased in 3D co-cultures of hMSCs with human umbilical vein endothelial cells (HUVECs). Additionally, we demonstrate VBP of a perfusable, pre-vascularized model where HUVECs self-organized into an endothelium-lined channel. Altogether, this work leverages the benefits of VBP and 3D co-culture, offering a promising platform for fast scaled biofabrication of 3D bone-like tissues with unprecedented functionality. STATEMENT OF SIGNIFICANCE: This study explores new strategies for ultrafast bio-manufacturing of bone tissue models by leveraging the advantages of tomographic volumetric bioprinting (VBP) and endothelial co-culture. After screening the properties of a series of photocurable gelatin methacryloyl (GelMA) bioresins, a formulation with 5% GelMA was identified with optimal printability and permissiveness for osteogenic differentiation of human mesenchymal stem cells (hMSC). We then established 3D endothelial co-cultures to test if the heterocellular interactions may enhance the osteogenic differentiation in the printed environments. This hypothesis was evidenced by increased gene expression of early osteocytic markers in 3D co-cultures after 21 days. Finally, VBP of a perfusable cell-laden tissue construct is demonstrated for future applications in vascularized tissue engineering.
Collapse
Affiliation(s)
- Jenny Gehlen
- Institute for Biomechanics, ETH Zürich, Leopold-Ruzicka-Weg 4, 8093 Zürich, Switzerland
| | - Wanwan Qiu
- Institute for Biomechanics, ETH Zürich, Leopold-Ruzicka-Weg 4, 8093 Zürich, Switzerland
| | - Gian Nutal Schädli
- Institute for Biomechanics, ETH Zürich, Leopold-Ruzicka-Weg 4, 8093 Zürich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zürich, Leopold-Ruzicka-Weg 4, 8093 Zürich, Switzerland
| | - Xiao-Hua Qin
- Institute for Biomechanics, ETH Zürich, Leopold-Ruzicka-Weg 4, 8093 Zürich, Switzerland.
| |
Collapse
|
8
|
de Wildt BWM, Cramer EEA, de Silva LS, Ito K, Gawlitta D, Hofmann S. Evaluating material-driven regeneration in a tissue engineered human in vitro bone defect model. Bone 2023; 166:116597. [PMID: 36280106 DOI: 10.1016/j.bone.2022.116597] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/07/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022]
Abstract
Advanced in vitro human bone defect models can contribute to the evaluation of materials for in situ bone regeneration, addressing both translational and ethical concerns regarding animal models. In this study, we attempted to develop such a model to study material-driven regeneration, using a tissue engineering approach. By co-culturing human umbilical vein endothelial cells (HUVECs) with human bone marrow-derived mesenchymal stromal cells (hBMSCs) on silk fibroin scaffolds with in vitro critically sized defects, the growth of vascular-like networks and three-dimensional bone-like tissue was facilitated. After a model build-up phase of 28 days, materials were artificially implanted and HUVEC and hBMSC migration, cell-material interactions, and osteoinduction were evaluated 14 days after implantation. The materials physiologically relevant for bone regeneration included a platelet gel as blood clot mimic, cartilage spheres as soft callus mimics, and a fibrin gel as control. Although the in vitro model was limited in the evaluation of immune responses, hallmarks of physiological bone regeneration were observed in vitro. These included the endothelial cell chemotaxis induced by the blood clot mimic and the mineralization of the soft callus mimic. Therefore, the present in vitro model could contribute to an improved pre-clinical evaluation of biomaterials while reducing the need for animal experiments.
Collapse
Affiliation(s)
- Bregje W M de Wildt
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Esther E A Cramer
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Leanne S de Silva
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
9
|
Geevarghese R, Sajjadi SS, Hudecki A, Sajjadi S, Jalal NR, Madrakian T, Ahmadi M, Włodarczyk-Biegun MK, Ghavami S, Likus W, Siemianowicz K, Łos MJ. Biodegradable and Non-Biodegradable Biomaterials and Their Effect on Cell Differentiation. Int J Mol Sci 2022; 23:ijms232416185. [PMID: 36555829 PMCID: PMC9785373 DOI: 10.3390/ijms232416185] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Biomaterials for tissue scaffolds are key components in modern tissue engineering and regenerative medicine. Targeted reconstructive therapies require a proper choice of biomaterial and an adequate choice of cells to be seeded on it. The introduction of stem cells, and the transdifferentiation procedures, into regenerative medicine opened a new era and created new challenges for modern biomaterials. They must not only fulfill the mechanical functions of a scaffold for implanted cells and represent the expected mechanical strength of the artificial tissue, but furthermore, they should also assure their survival and, if possible, affect their desired way of differentiation. This paper aims to review how modern biomaterials, including synthetic (i.e., polylactic acid, polyurethane, polyvinyl alcohol, polyethylene terephthalate, ceramics) and natural (i.e., silk fibroin, decellularized scaffolds), both non-biodegradable and biodegradable, could influence (tissue) stem cells fate, regulate and direct their differentiation into desired target somatic cells.
Collapse
Affiliation(s)
- Rency Geevarghese
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Seyedeh Sara Sajjadi
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
| | - Andrzej Hudecki
- Łukasiewicz Network-Institute of Non-Ferrous Metals, 44-121 Gliwice, Poland
| | - Samad Sajjadi
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
| | | | - Tayyebeh Madrakian
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6516738695, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mazaher Ahmadi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6516738695, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Małgorzata K. Włodarczyk-Biegun
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Saeid Ghavami
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
| | - Wirginia Likus
- Department of Anatomy, Faculty of Health Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
| | - Krzysztof Siemianowicz
- Department of Biochemistry, Faculty of Medicine in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
- Correspondence: (K.S.); (M.J.Ł.); Tel.: +48-32-237-2913 (M.J.Ł.)
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Correspondence: (K.S.); (M.J.Ł.); Tel.: +48-32-237-2913 (M.J.Ł.)
| |
Collapse
|
10
|
Bouten CVC, Cheng C, Vermue IM, Gawlitta D, Passier R. Cardiovascular tissue engineering and regeneration: A plead for further knowledge convergence. Tissue Eng Part A 2022; 28:525-541. [PMID: 35382591 DOI: 10.1089/ten.tea.2021.0231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular tissue engineering and regeneration strive to provide long-term, effective solutions for a growing group of patients in need of myocardial repair, vascular (access) grafts, heart valves, and regeneration of organ microcirculation. In the past two decades, ongoing convergence of disciplines and multidisciplinary collaborations between cardiothoracic surgeons, cardiologists, bioengineers, material scientists, and cell biologists have resulted in better understanding of the problems at hand and novel regenerative approaches. As a side effect, however, the field has become strongly organized and differentiated around topical areas at risk of reinvention of technologies and repetition of approaches and across the areas. A better integration of knowledge and technologies from the individual topical areas and regenerative approaches and technologies may pave the way towards faster and more effective treatments to cure the cardiovascular system. This review summarizes the evolution of research and regenerative approaches in the areas of myocardial regeneration, heart valve and vascular tissue engineering, and regeneration of microcirculations and discusses previous and potential future integration of these individual areas and developed technologies for improved clinical impact. Finally, it provides a perspective on the further integration of research organization, knowledge implementation, and valorization as a contributor to advancing cardiovascular tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Carlijn V C Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Caroline Cheng
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ijsbrand M Vermue
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery, Prosthodontics and Special Dental Care, University Medical Center, Utrecht, The Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
11
|
Lin Z, Zhang X, Fritch MR, Li Z, Kuang B, Alexander PG, Hao T, Cao G, Tan S, Bruce KK, Lin H. Engineering pre-vascularized bone-like tissue from human mesenchymal stem cells through simulating endochondral ossification. Biomaterials 2022; 283:121451. [DOI: 10.1016/j.biomaterials.2022.121451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/28/2022] [Accepted: 02/27/2022] [Indexed: 01/12/2023]
|
12
|
ABSTRACTS (BY NUMBER). Tissue Eng Part A 2022. [DOI: 10.1089/ten.tea.2022.29025.abstracts] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
13
|
Li J, Cui X, Lindberg G, Alcala-Orozco CR, Hooper GJ, Lim K, Woodfield TBF. Hybrid fabrication of photo-clickable vascular hydrogels with additive manufactured titanium implants for enhanced osseointegration and vascularized bone formation. Biofabrication 2022; 14. [PMID: 35320796 DOI: 10.1088/1758-5090/ac6051] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/23/2022] [Indexed: 11/11/2022]
Abstract
Bone regeneration of critical-sized bone defects, bone fractures or joint replacements remains a significant unmet clinical challenge. Although there has been rapid advancement in both the fields of bone tissue engineering and additive manufacturing (AM), functional bone implants with rapid vascularization capacity to ensure osseointegration and long-term biological fixation in large bone defects remains limited in clinics. In this study, we developed an in vitro vascularized bone implant by combining cell-laden hydrogels with direct metal printed (DMP) porous titanium alloys (Ti-6Al-4V). 5wt% allylated gelatin (GelAGE), was utilized to co-encapsulate human mesenchymal stromal cells (hMSCs) and human umbilical vein endothelial cells (HUVECs) to investigate concurrent osteogenic and vasculogenic performance. DMP macro-porous Ti-6Al-4V scaffolds were subsequently infused/enriched with cell-laden GelAGE to examine the feasibility to deliver cells and engineer vascular-like networks in the hybrid implant. Furthermore, as a proof of concept, a full-scale porous Ti-6Al-4V acetabular cup was impregnated with cell-laden hydrogel to validate the clinical potential of this strategy. The vasculogenic potential was evaluated by examining micro-capillary formation coupled with capillary network maturation and stabilization. Osteogenic differentiation was assessed via ALP activity as well as osteocalcin and osteopontin expression. Our results suggested that GelAGE supported HUVECs spreading and vascular-like network formation, along with osteogenesis of hMSCs. Titanium hybrid constructs with cell-laden hydrogel demonstrated enhanced osteogenesis with similar vasculogenic capability compared to the cell-laden hydrogel alone constructs. The full-scale implant with cell-laden hydrogel coating similarly showed cell distribution and spreading, implying the potential for further clinical application. Our study presents the feasibility of integrating bio-functional hydrogels with porous titanium implants to fabricate a vascularized hybrid construct with both mechanical support and preferable biological functionality (osteogenesis/vasculogenesis), which paves the way for improved strategies to enhance bone regeneration in complex large bone defects achieving long-term bone-implant fixation.
Collapse
Affiliation(s)
- Jun Li
- Dept. of Orthopaedic Surgery , University of Otago, 2 Riccarton Avenue, Christchurch, Christchurch, Canterbury, 8011, NEW ZEALAND
| | - Xiaolin Cui
- University of Otago Christchurch, 2 Riccarton Avenue, Christchurch, 8011, NEW ZEALAND
| | - Gabriella Lindberg
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, 2 Riccarton Avenue, Christchurch, 8011, NEW ZEALAND
| | - Cesar R Alcala-Orozco
- Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, 2 Riccarton Avenue, Christchurch, Christchurch, 8011, NEW ZEALAND
| | - Gary J Hooper
- Christchurch Regenerative Medicine and Tissue Engineering Group Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, PO Box 4345, Christchurch 8140, Christchurch, 8140, NEW ZEALAND
| | - Khoon Lim
- Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, 2 Riccarton Avenue, Christchurch, 8011, NEW ZEALAND
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering Group Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, PO Box 4345, Christchurch 8140, Christchurch, 8140, NEW ZEALAND
| |
Collapse
|
14
|
Camman M, Joanne P, Agbulut O, Hélary C. 3D models of dilated cardiomyopathy: Shaping the chemical, physical and topographical properties of biomaterials to mimic the cardiac extracellular matrix. Bioact Mater 2022; 7:275-291. [PMID: 34466733 PMCID: PMC8379361 DOI: 10.1016/j.bioactmat.2021.05.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
The pathophysiology of dilated cardiomyopathy (DCM), one major cause of heart failure, is characterized by the dilation of the heart but remains poorly understood because of the lack of adequate in vitro models. Current 2D models do not allow for the 3D organotypic organization of cardiomyocytes and do not reproduce the ECM perturbations. In this review, the different strategies to mimic the chemical, physical and topographical properties of the cardiac tissue affected by DCM are presented. The advantages and drawbacks of techniques generating anisotropy required for the cardiomyocytes alignment are discussed. In addition, the different methods creating macroporosity and favoring organotypic organization are compared. Besides, the advances in the induced pluripotent stem cells technology to generate cardiac cells from healthy or DCM patients will be described. Thanks to the biomaterial design, some features of the DCM extracellular matrix such as stiffness, porosity, topography or chemical changes can impact the cardiomyocytes function in vitro and increase their maturation. By mimicking the affected heart, both at the cellular and at the tissue level, 3D models will enable a better understanding of the pathology and favor the discovery of novel therapies.
Collapse
Affiliation(s)
- Marie Camman
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Pierre Joanne
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
| |
Collapse
|
15
|
Soliman BG, Major GS, Atienza-Roca P, Murphy CA, Longoni A, Alcala-Orozco CR, Rnjak-Kovacina J, Gawlitta D, Woodfield TBF, Lim KS. Development and Characterization of Gelatin-Norbornene Bioink to Understand the Interplay between Physical Architecture and Micro-Capillary Formation in Biofabricated Vascularized Constructs. Adv Healthc Mater 2022; 11:e2101873. [PMID: 34710291 DOI: 10.1002/adhm.202101873] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/21/2021] [Indexed: 12/12/2022]
Abstract
The principle challenge for engineering viable, cell-laden hydrogel constructs of clinically-relevant size, is rapid vascularization, in order to moderate the finite capacity of passive nutrient diffusion. A multiscale vascular approach, with large open channels and bulk microcapillaries may be an admissible approach to accelerate this process, promoting overall pre-vascularization for long-term viability of constructs. However, the limited availability of bioinks that possess suitable characteristics that support both fabrication of complex architectures and formation of microcapillaries, remains a barrier to advancement in this space. In this study, gelatin-norbornene (Gel-NOR) is investigated as a vascular bioink with tailorable physico-mechanical properties, which promoted the self-assembly of human stromal and endothelial cells into microcapillaries, as well as being compatible with extrusion and lithography-based biofabrication modalities. Gel-NOR constructs containing self-assembled microcapillaries are successfully biofabricated with varying physical architecture (fiber diameter, spacing, and orientation). Both channel sizes and cell types affect the overall structural changes of the printed constructs, where cross-signaling between both human stromal and endothelial cells may be responsible for the reduction in open channel lumen observed over time. Overall, this work highlights an exciting three-way interplay between bioink formulation, construct design, and cell-mediated response that can be exploited towards engineering vascular tissues.
Collapse
Affiliation(s)
- Bram G Soliman
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Gretel S Major
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Pau Atienza-Roca
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Caroline A Murphy
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Alessia Longoni
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Cesar R Alcala-Orozco
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2006, Australia
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery and Special Dental Care, University Medical Center Utrecht, Utrecht, GA, 3508, The Netherlands
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Khoon S Lim
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| |
Collapse
|
16
|
Xu HZ, Su JS. Restoration of critical defects in the rabbit mandible using osteoblasts and vascular endothelial cells co-cultured with vascular stent-loaded nano-composite scaffolds. J Mech Behav Biomed Mater 2021; 124:104831. [PMID: 34555626 DOI: 10.1016/j.jmbbm.2021.104831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 01/07/2023]
Abstract
The success of large bone defect repair with tissue engineering technology depends mainly on angiogenesis and osteogenesis. In this study, we prepared poly-caprolactone/nano-hydroxyapatite/beta-calcium phosphate (PCL/nHA/β-TCP) composite scaffolds loaded with poly-(lactic-co-glycolic acid)/nano-hydroxyapatite/collagen/heparin sodium (PLGA/nHA/Col/HS) nanofiber small vascular stent by electrospinning and hot press forming-particle leaching methods. Supramolecular electrostatic self-assembly technology was used to modify the surfaces of small vascular stents to aid in hydrophilicity and anticoagulation. The surfaces of composite scaffolds were modified with an Arg-Gly-Asp (RGD) short peptide by physical adsorption to supply cell adhesion sites. The scaffolds were then combined with rabbit bone marrow-derived osteoblasts (OBs) and rabbit bone marrow-derived vascular endothelial cells (RVECs) to construct large, biologically active vascularized tissue-engineered bone in vitro; this bone was then used to repair critical bone defects in rabbit mandibles. Mechanical and biocompatibility testing results showed that PCL/nHA/β-TCP composite scaffolds loaded with small vascular stents had good surface structure, mechanical properties, biocompatibility, and bone-regeneration induction potential. Twelve weeks after implantation, histological analysis and X-ray scans showed that the use of osteoblasts and vascular endothelial cells co-cultured with PCL/nHA/β-TCP scaffolds was sufficient to repair critical defects in rabbit mandibles.
Collapse
Affiliation(s)
- Hong Zhen Xu
- Department of Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Jian Sheng Su
- Department of Prosthodontics, School & Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China.
| |
Collapse
|
17
|
De Moor L, Smet J, Plovyt M, Bekaert B, Vercruysse C, Asadian M, De Geyter N, Van Vlierberghe S, Dubruel P, Declercq H. Engineering microvasculature by 3D bioprinting of prevascularized spheroids in photo-crosslinkable gelatin. Biofabrication 2021; 13. [PMID: 34496350 DOI: 10.1088/1758-5090/ac24de] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/08/2021] [Indexed: 01/01/2023]
Abstract
To engineer tissues with clinically relevant dimensions by three-dimensional bioprinting, an extended vascular network with diameters ranging from the macro- to micro-scale needs to be integrated. Extrusion-based bioprinting is the most commonly applied bioprinting technique but due to the limited resolution of conventional bioprinters, the establishment of a microvascular network for the transfer of oxygen, nutrients and metabolic waste products remains challenging. To answer this need, this study assessed the potential and processability of spheroids, containing a capillary-like network, to be used as micron-sized prevascularized units for incorporation throughout the bioprinted construct. Prevascularized spheroids were generated by combining endothelial cells with fibroblasts and adipose tissue-derived mesenchymal stem cells as supporting cells. To serve as a viscous medium for the bioink-based deposition by extrusion printing, spheroids were combined with a photo-crosslinkable methacrylamide-modified gelatin (gelMA) and Irgacure 2959. The influence of gelMA encapsulation, the printing process and photo-crosslinking conditions on spheroid viability, proliferation and vascularization were analyzed by live/dead staining, immunohistochemistry, gene expression analysis and sprouting analysis. Stable spheroid-laden constructs, allowing spheroid outgrowth, were achieved by applying 10 min UV-A photo-curing (365 nm, 4 mW cm-2), while the construct was incubated in an additional Irgacure 2959 immersion solution. Following implantationin ovoonto a chick chorioallantoic membrane, the prevascular engineered constructs showed anastomosis with the host vasculature. This study demonstrated (a) the potential of triculture prevascularized spheroids for application as multicellular building blocks, (b) the processability of the spheroid-laden gelMA bioink by extrusion bioprinting and (c) the importance of photo-crosslinking parameters post printing, as prolonged photo-curing intervals showed to be detrimental for the angiogenic potential and complete vascularization of the construct post printing.
Collapse
Affiliation(s)
- Lise De Moor
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Jasper Smet
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.,Tissue Engineering Lab, Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Kortrijk, Belgium
| | - Magalie Plovyt
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Bieke Bekaert
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Chris Vercruysse
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Mahtab Asadian
- Research Unit Plasma Technology, Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Ghent, Belgium
| | - Nathalie De Geyter
- Research Unit Plasma Technology, Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Research Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Research Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Heidi Declercq
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.,Tissue Engineering Lab, Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Kortrijk, Belgium
| |
Collapse
|
18
|
Alcala-Orozco CR, Cui X, Hooper GJ, Lim KS, Woodfield TB. Converging functionality: Strategies for 3D hybrid-construct biofabrication and the role of composite biomaterials for skeletal regeneration. Acta Biomater 2021; 132:188-216. [PMID: 33713862 DOI: 10.1016/j.actbio.2021.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/02/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022]
Abstract
The evolution of additive manufacturing (AM) technologies, biomaterial development and our increasing understanding of cell biology has created enormous potential for the development of personalized regenerative therapies. In the context of skeletal tissue engineering, physical and biological demands play key roles towards successful construct implantation and the achievement of bone, cartilage and blood vessel tissue formation. Nevertheless, meeting such physical and biological demands to mimic the complexity of human tissues and their functionality is still a significant ongoing challenge. Recent studies have demonstrated that combination of AM technologies and advanced biomaterials has great potential towards skeletal tissue engineering. This review aims to analyze how the most prominent technologies and discoveries in the field converge towards the development of advanced constructs for skeletal regeneration. Particular attention is placed on hybrid biofabrication strategies, combining bioinks for cell delivery with biomaterial inks providing physical support. Hybrid biofabrication has been the focus of recent emerging strategies, however there has been limited review and analysis of these techniques and the challenges involved. Furthermore, we have identified that there are multiple hybrid fabrication strategies, here we present a category system where each strategy is reviewed highlighting their distinct advantages, challenges and potential applications. In addition, bioinks and biomaterial inks are the main components of the hybrid biofabrication strategies, where it is recognized that such platforms still lack optimal physical and biological functionality. Thus, this review also explores the development of composite materials specifically targeting the enhancement of physical and biological functionality towards improved skeletal tissue engineering. STATEMENT OF SIGNIFICANCE: Biofabrication strategies capable of recreating the complexity of native tissues could open new clinical possibilities towards patient-specific regenerative therapies and disease models. Several reviews target the existing additive manufacturing (AM) technologies that may be utilised for biomedical purposes. However, this work presents a unique perspective, describing how such AM technologies have been recently translated towards hybrid fabrication strategies, targeting the fabrication of constructs with converging physical and biological properties. Furthermore, we address composite bioinks and biomaterial inks that have been engineered to overcome traditional limitations, and might be applied to the hybrid fabrication strategies outlined. This work offers ample perspectives and insights into the current and future challenges for the fabrication of skeletal tissues aiming towards clinical and biomedical applications.
Collapse
|
19
|
Levato R, Lim KS, Li W, Asua AU, Peña LB, Wang M, Falandt M, Bernal PN, Gawlitta D, Zhang YS, Woodfield TBF, Malda J. High-resolution lithographic biofabrication of hydrogels with complex microchannels from low-temperature-soluble gelatin bioresins. Mater Today Bio 2021; 12:100162. [PMID: 34870141 PMCID: PMC8626672 DOI: 10.1016/j.mtbio.2021.100162] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022] Open
Abstract
Biofabrication via light-based 3D printing offers superior resolution and ability to generate free-form architectures, compared to conventional extrusion technologies. While extensive efforts in the design of new hydrogel bioinks lead to major advances in extrusion methods, the accessibility of lithographic bioprinting is still hampered by a limited choice of cell-friendly resins. Herein, we report the development of a novel set of photoresponsive bioresins derived from ichthyic-origin gelatin, designed to print high-resolution hydrogel constructs with embedded convoluted networks of vessel-mimetic channels. Unlike mammalian gelatins, these materials display thermal stability as pre-hydrogel solutions at room temperature, ideal for bioprinting on any easily-accessible lithographic printer. Norbornene- and methacryloyl-modification of the gelatin backbone, combined with a ruthenium-based visible light photoinitiator and new coccine as a cytocompatible photoabsorber, allowed to print structures resolving single-pixel features (∼50 μm) with high shape fidelity, even when using low stiffness gels, ideal for cell encapsulation (1-2 kPa). Moreover, aqueous two-phase emulsion bioresins allowed to modulate the permeability of the printed hydrogel bulk. Bioprinted mesenchymal stromal cells displayed high functionality over a month of culture, and underwent multi-lineage differentiation while colonizing the bioresin bulk with tissue-specific neo-deposited extracellular matrix. Importantly, printed hydrogels embedding complex channels with perfusable lumen (diameter <200 μm) were obtained, replicating anatomical 3D networks with out-of-plane branches (i.e. brain vessels) that cannot otherwise be reproduced by extrusion bioprinting. This versatile bioresin platform opens new avenues for the widespread adoption of lithographic biofabrication, and for bioprinting complex channel-laden constructs with envisioned applications in regenerative medicine and hydrogel-based organ-on-a-chip devices.
Collapse
Affiliation(s)
- Riccardo Levato
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
- Department of Orthopaedics, University Medical Center Utrecht, the Netherlands
| | - Khoon S Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, the Netherlands
| | - Wanlu Li
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, USA
| | - Ane Urigoitia Asua
- Department of Orthopaedics, University Medical Center Utrecht, the Netherlands
| | - Laura Blanco Peña
- Department of Orthopaedics, University Medical Center Utrecht, the Netherlands
| | - Mian Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, USA
| | - Marc Falandt
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
| | | | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, the Netherlands
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, USA
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, the Netherlands
| | - Jos Malda
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
- Department of Orthopaedics, University Medical Center Utrecht, the Netherlands
| |
Collapse
|
20
|
Strategies for inclusion of growth factors into 3D printed bone grafts. Essays Biochem 2021; 65:569-585. [PMID: 34156062 DOI: 10.1042/ebc20200130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
There remains a critical need to develop new technologies and materials that can meet the demands of treating large bone defects. The advancement of 3-dimensional (3D) printing technologies has allowed the creation of personalized and customized bone grafts, with specific control in both macro- and micro-architecture, and desired mechanical properties. Nevertheless, the biomaterials used for the production of these bone grafts often possess poor biological properties. The incorporation of growth factors (GFs), which are the natural orchestrators of the physiological healing process, into 3D printed bone grafts, represents a promising strategy to achieve the bioactivity required to enhance bone regeneration. In this review, the possible strategies used to incorporate GFs to 3D printed constructs are presented with a specific focus on bone regeneration. In particular, the strengths and limitations of different methods, such as physical and chemical cross-linking, which are currently used to incorporate GFs to the engineered constructs are critically reviewed. Different strategies used to present one or more GFs to achieve simultaneous angiogenesis and vasculogenesis for enhanced bone regeneration are also covered in this review. In addition, the possibility of combining several manufacturing approaches to fabricate hybrid constructs, which better mimic the complexity of biological niches, is presented. Finally, the clinical relevance of these approaches and the future steps that should be taken are discussed.
Collapse
|
21
|
Lee H, Kim DI, Kwon SH, Park S. Magnetically Actuated Drug Delivery Helical Microrobot with Magnetic Nanoparticle Retrieval Ability. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19633-19647. [PMID: 33877809 DOI: 10.1021/acsami.1c01742] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Therapeutic drug delivery microrobots capable of accurate targeting using an electromagnetic actuation (EMA) system are being developed. However, these drug delivery microrobots include a large number of magnetic nanoparticles (MNPs) for accurate EMA targeting, which causes side effects, such as problems with membrane integrity and normal cell apoptosis. Here, a biocompatible and hydrolyzable PEGDA-based drug delivery helical microrobot capable of MNP retrieval is proposed in which doxorubicin (DOX), an anticancer drug, is encapsulated and MNPs are conjugated by a disulfide bond. After being accurately delivered to the lesion of cancer cells through magnetic field manipulation, the fabricated microrobot provides rapid MNP separation and retrieval from the microrobot because of the use of dithiothreitol (DTT), a reducing agent, as an environment similar to the surrounding cancer cells and near-infrared (NIR) as an external stimulus. The characteristics of the fabricated microrobot are analyzed, and fundamental tests for active electromagnetic field manipulation, separation/retrieval of MNPs from the microrobot, and its hydrolysis are discussed. The therapeutic performance of the fabricated microrobot is verified through an in vitro test using tumor cells. Consequently, by use of an integrated system of microscope, eight-coil EMA, and NIR it is shown that the proposed microrobot can be moved to the target site by electromagnetic manipulation. The MNPs conjugated to the microrobot can be separated and retrieved, and the therapeutic effect on tumor cells by the encapsulated drug can be seen.
Collapse
Affiliation(s)
- Hyoryong Lee
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Dong-In Kim
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Su-Hyun Kwon
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Sukho Park
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| |
Collapse
|
22
|
Nulty J, Freeman FE, Browe DC, Burdis R, Ahern DP, Pitacco P, Lee YB, Alsberg E, Kelly DJ. 3D bioprinting of prevascularised implants for the repair of critically-sized bone defects. Acta Biomater 2021; 126:154-169. [PMID: 33705989 DOI: 10.1016/j.actbio.2021.03.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/06/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
For 3D bioprinted tissues to be scaled-up to clinically relevant sizes, effective prevascularisation strategies are required to provide the necessary nutrients for normal metabolism and to remove associated waste by-products. The aim of this study was to develop a bioprinting strategy to engineer prevascularised tissues in vitro and to investigate the capacity of such constructs to enhance the vascularisation and regeneration of large bone defects in vivo. From a screen of different bioinks, a fibrin-based hydrogel was found to best support human umbilical vein endothelial cell (HUVEC) sprouting and the establishment of a microvessel network. When this bioink was combined with HUVECs and supporting human bone marrow stem/stromal cells (hBMSCs), these microvessel networks persisted in vitro. Furthermore, only bioprinted tissues containing both HUVECs and hBMSCs, that were first allowed to mature in vitro, supported robust blood vessel development in vivo. To assess the therapeutic utility of this bioprinting strategy, these bioinks were used to prevascularise 3D printed polycaprolactone (PCL) scaffolds, which were subsequently implanted into critically-sized femoral bone defects in rats. Micro-computed tomography (µCT) angiography revealed increased levels of vascularisation in vivo, which correlated with higher levels of new bone formation. Such prevascularised constructs could be used to enhance the vascularisation of a range of large tissue defects, forming the basis of multiple new bioprinted therapeutics. STATEMENT OF SIGNIFICANCE: This paper demonstrates a versatile 3D bioprinting technique to improve the vascularisation of tissue engineered constructs and further demonstrates how this method can be incorporated into a bone tissue engineering strategy to improve vascularisation in a rat femoral defect model.
Collapse
|
23
|
Vascular bioprinting with enzymatically degradable bioinks via multi-material projection-based stereolithography. Acta Biomater 2020; 117:121-132. [PMID: 32980542 DOI: 10.1016/j.actbio.2020.09.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/02/2020] [Accepted: 09/18/2020] [Indexed: 12/20/2022]
Abstract
Introduction of cavities and channels into 3D bioprinted constructs is a prerequisite for recreating physiological tissue architectures and integrating vasculature. Projection-based stereolithography inherently offers high printing speed with high spatial resolution, but so far has been incapable of fabricating complex native tissue architectures with cellular and biomaterial diversity. The use of sacrificial photoinks, i.e. photopolymerisable biomaterials that can be removed after printing, theoretically allows for the creation of any construct geometry via a multi-material printing process. However, the realisation of this strategy has been challenging because of difficult technical implementation and a lack of performant biomaterials. In this work, we use our projection-based, multi-material stereolithographic bioprinter and an enzymatically degradable sacrificial photoink to overcome the current hurdles. Multiple, hyaluronic acid-based photoinks were screened for printability, mechanical properties and digestibility through hyaluronidase. A formulation meeting all major requirements, i.e. desirable printing properties, generation of sufficiently strong hydrogels, as well as fast digestion rate, was identified. Biocompatibility of the material system was confirmed by embedding of human umbilical vein endothelial cells with followed enzymatic release. As a proof-of-concept, we bioprinted vascular models containing perfusable, endothelial cell-lined channels that remained stable for 28 days in culture. Our work establishes digestible sacrificial biomaterials as a new material strategy for 3D bioprinting of complex tissue models.
Collapse
|
24
|
Kreuder AE, Bolaños-Rosales A, Palmer C, Thomas A, Geiger MA, Lam T, Amler AK, Markert UR, Lauster R, Kloke L. Inspired by the human placenta: a novel 3D bioprinted membrane system to create barrier models. Sci Rep 2020; 10:15606. [PMID: 32973223 PMCID: PMC7515925 DOI: 10.1038/s41598-020-72559-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 08/28/2020] [Indexed: 12/15/2022] Open
Abstract
Barrier organ models need a scaffold structure to create a two compartment culture. Technical filter membranes used most often as scaffolds may impact cell behaviour and present a barrier themselves, ultimately limiting transferability of test results. In this work we present an alternative for technical filter membrane systems: a 3D bioprinted biological membrane in 24 well format. The biological membrane, based on extracellular matrix (ECM), is highly permeable and presents a natural 3D environment for cell culture. Inspired by the human placenta we established a coculture of a trophoblast-derived cell line (BeWo b30), together with primary placental fibroblasts within the biological membrane (simulating villous stroma) and primary human placental endothelial cells-representing three cellular components of the human placental villus. All cell types maintained their cell type specific marker expression after two weeks of coculture on the biological membrane. In permeability assays the trophoblast layer developed a barrier on the biological membrane, which was even more pronounced when cocultured with fibroblasts. In this work we present a filter membrane free scaffold, we characterize its properties and assess its suitability for cell culture and barrier models. Further we show a novel placenta inspired model in a complex bioprinted coculture. In the absence of an artificial filter membrane, we demonstrate barrier architecture and functionality.
Collapse
Affiliation(s)
- Anna-Elisabeth Kreuder
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany.
- Cellbricks GmbH, Berlin, 13355, Germany.
| | - Aramis Bolaños-Rosales
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany
- Cellbricks GmbH, Berlin, 13355, Germany
| | | | - Alexander Thomas
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany
- Cellbricks GmbH, Berlin, 13355, Germany
| | | | | | - Anna-Klara Amler
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany
- Cellbricks GmbH, Berlin, 13355, Germany
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, University Hospital Jena, 07747, Jena, Germany
| | - Roland Lauster
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany
| | - Lutz Kloke
- Cellbricks GmbH, Berlin, 13355, Germany.
| |
Collapse
|
25
|
Soliman BG, Lindberg GCJ, Jungst T, Hooper GJ, Groll J, Woodfield TBF, Lim KS. Stepwise Control of Crosslinking in a One-Pot System for Bioprinting of Low-Density Bioinks. Adv Healthc Mater 2020; 9:e1901544. [PMID: 32323473 DOI: 10.1002/adhm.201901544] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022]
Abstract
Extrusion-based 3D bioprinting is hampered by the inability to print materials of low-viscosity. In this study, a single initiating system based on ruthenium (Ru) and sodium persulfate (SPS) is utilized for a sequential dual-step crosslinking approach: 1) primary (partial) crosslinking in absence of light to alter the bioink's rheological profile for print fidelity, and 2) subsequent secondary post-printing crosslinking for shape maintenance. Allyl-functionalized gelatin (Gel-AGE) is used as a bioink, allowing thiol-ene click reaction between allyl moieties and thiolated crosslinkers. A systematic investigation of primary crosslinking reveals that a thiol-persulfate redox reaction facilitates thiol-ene crosslinking, mediating an increase in bioink viscosity that is controllable by tailoring the Ru/SPS, crosslinker, and/or Gel-AGE concentrations. Thereafter, subsequent photoinitiated secondary crosslinking then facilitates maximum conversion of thiol-ene bonds between AGE and thiol groups. The dual-step crosslinking method is applicable to a wide biofabrication window (3-10 wt% Gel-AGE) and is demonstrated to allow printing of low-density (3 wt%) Gel-AGE, normally exhibiting low viscosity (4 mPa s), with high shape fidelity and high cell viability (>80%) over 7 days of culture. The presented approach can therefore be used as a one-pot system for printing low-viscous bioinks without the need for multiple initiating systems, viscosity enhancers, or complex chemical modifications.
Collapse
Affiliation(s)
- Bram G. Soliman
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupUniversity of Otago 2 Riccarton Avenue Christchurch 8011 New Zealand
| | - Gabriella C. J. Lindberg
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupUniversity of Otago 2 Riccarton Avenue Christchurch 8011 New Zealand
- Medical Technologies Centre of Research Excellence Auckland 1010 New Zealand
| | - Tomasz Jungst
- Department for Functional Materials in Medicine and Dentistry (FMZ) and Bavarian Polymer Institute (BPI)University of Würzburg Pleicherwall 2 Würzburg 97070 Germany
| | - Gary J. Hooper
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupUniversity of Otago 2 Riccarton Avenue Christchurch 8011 New Zealand
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry (FMZ) and Bavarian Polymer Institute (BPI)University of Würzburg Pleicherwall 2 Würzburg 97070 Germany
| | - Tim B. F. Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupUniversity of Otago 2 Riccarton Avenue Christchurch 8011 New Zealand
- Medical Technologies Centre of Research Excellence Auckland 1010 New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery Auckland 1010 New Zealand
| | - Khoon S. Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupUniversity of Otago 2 Riccarton Avenue Christchurch 8011 New Zealand
- Medical Technologies Centre of Research Excellence Auckland 1010 New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery Auckland 1010 New Zealand
| |
Collapse
|
26
|
Cui X, Li J, Hartanto Y, Durham M, Tang J, Zhang H, Hooper G, Lim K, Woodfield T. Advances in Extrusion 3D Bioprinting: A Focus on Multicomponent Hydrogel-Based Bioinks. Adv Healthc Mater 2020; 9:e1901648. [PMID: 32352649 DOI: 10.1002/adhm.201901648] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/14/2020] [Accepted: 03/17/2020] [Indexed: 12/18/2022]
Abstract
3D bioprinting involves the combination of 3D printing technologies with cells, growth factors and biomaterials, and has been considered as one of the most advanced tools for tissue engineering and regenerative medicine (TERM). However, despite multiple breakthroughs, it is evident that numerous challenges need to be overcome before 3D bioprinting will eventually become a clinical solution for a variety of TERM applications. To produce a 3D structure that is biologically functional, cell-laden bioinks must be optimized to meet certain key characteristics including rheological properties, physico-mechanical properties, and biofunctionality; a difficult task for a single component bioink especially for extrusion based bioprinting. As such, more recent research has been centred on multicomponent bioinks consisting of a combination of two or more biomaterials to improve printability, shape fidelity and biofunctionality. In this article, multicomponent hydrogel-based bioink systems are systemically reviewed based on the inherent nature of the bioink (natural or synthetic hydrogels), including the most current examples demonstrating properties and advances in application of multicomponent bioinks, specifically for extrusion based 3D bioprinting. This review article will assist researchers in the field in identifying the most suitable bioink based on their requirements, as well as pinpointing current unmet challenges in the field.
Collapse
Affiliation(s)
- Xiaolin Cui
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
- Medical Technologies Centre of Research Excellence, Auckland, 1142, New Zealand
| | - Jun Li
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Yusak Hartanto
- Department of Chemical Engineering, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Mitchell Durham
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Junnan Tang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Hu Zhang
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Gary Hooper
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
- Medical Technologies Centre of Research Excellence, Auckland, 1142, New Zealand
| | - Khoon Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
- Medical Technologies Centre of Research Excellence, Auckland, 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, 1142, New Zealand
| | - Tim Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
- Medical Technologies Centre of Research Excellence, Auckland, 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, 1142, New Zealand
| |
Collapse
|
27
|
Alcala-Orozco CR, Mutreja I, Cui X, Kumar D, Hooper GJ, Lim KS, Woodfield TB. Design and characterisation of multi-functional strontium-gelatin nanocomposite bioinks with improved print fidelity and osteogenic capacity. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2019.e00073] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Levato R, Jungst T, Scheuring RG, Blunk T, Groll J, Malda J. From Shape to Function: The Next Step in Bioprinting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1906423. [PMID: 32045053 PMCID: PMC7116209 DOI: 10.1002/adma.201906423] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/08/2019] [Indexed: 05/04/2023]
Abstract
In 2013, the "biofabrication window" was introduced to reflect the processing challenge for the fields of biofabrication and bioprinting. At that time, the lack of printable materials that could serve as cell-laden bioinks, as well as the limitations of printing and assembly methods, presented a major constraint. However, recent developments have now resulted in the availability of a plethora of bioinks, new printing approaches, and the technological advancement of established techniques. Nevertheless, it remains largely unknown which materials and technical parameters are essential for the fabrication of intrinsically hierarchical cell-material constructs that truly mimic biologically functional tissue. In order to achieve this, it is urged that the field now shift its focus from materials and technologies toward the biological development of the resulting constructs. Therefore, herein, the recent material and technological advances since the introduction of the biofabrication window are briefly summarized, i.e., approaches how to generate shape, to then focus the discussion on how to acquire the biological function within this context. In particular, a vision of how biological function can evolve from the possibility to determine shape is outlined.
Collapse
Affiliation(s)
- Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Tomasz Jungst
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Ruben G Scheuring
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Juergen Groll
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|
29
|
Chiesa I, De Maria C, Lapomarda A, Fortunato GM, Montemurro F, Di Gesù R, Tuan RS, Vozzi G, Gottardi R. Endothelial cells support osteogenesis in an in vitro vascularized bone model developed by 3D bioprinting. Biofabrication 2020; 12:025013. [PMID: 31929117 DOI: 10.1088/1758-5090/ab6a1d] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bone is a highly vascularized tissue, in which vascularization and mineralization are concurrent processes during skeletal development. Indeed, both components should be included in any reliable and adherent in vitro model platform for the study of bone physiology and pathogenesis of skeletal disorders. To this end, we developed an in vitro vascularized bone model, using a gelatin-nanohydroxyapatite (gel-nHA) three-dimensional (3D) bioprinted scaffold. First, we seeded human mesenchymal stem cells (hMSCs) on the scaffold, which underwent osteogenic differentiation for 2 weeks. Then, we included lentiviral-GFP transfected human umbilical vein endothelial cells (HUVECs) within the 3D bioprinted scaffold macropores to form a capillary-like network during 2 more weeks of culture. We tested three experimental conditions: condition 1, bone constructs with HUVECs cultured in 1:1 osteogenic medium (OM): endothelial medium (EM); condition 2, bone constructs without HUVECs cultured in 1:1 OM:EM; condition 3: bone construct with HUVECs cultured in 1:1 growth medium:EM. All samples resulted in engineered bone matrix. In conditions 1 and 3, HUVECs formed tubular structures within the bone constructs, with the assembly of a complex capillary-like network visible by fluorescence microscopy in the live tissue and histology. CD31 immunostaining confirmed significant vascular lumen formation. Quantitative real-time PCR was used to quantify osteogenic differentiation and endothelial response. Alkaline phosphatase and runt-related transcription factor 2 upregulation confirmed early osteogenic commitment of hMSCs. Even when OM was removed under condition 3, we observed clear osteogenesis, which was notably accompanied by upregulation of osteopontin, vascular endothelial growth factor, and collagen type I. These findings indicate that we have successfully realized a bone model with robust vascularization in just 4 weeks of culture and we highlighted how the inclusion of endothelial cells more realistically supports osteogenesis. The approach reported here resulted in a biologically inspired in vitro model of bone vascularization, simulating de novo morphogenesis of capillary vessels occurring during tissue development.
Collapse
Affiliation(s)
- Irene Chiesa
- Research Center 'E. Piaggio', University of Pisa, Pisa, Italy. Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, United States of America. Department of Pediatrics, Division of Pulmonary Medicine, The Children's Hospital of Philadelphia, Philadelphia, United States of America. Dept. of Ingegneria dell'Informazione, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Pennings I, van Haaften EE, Jungst T, Bulsink JA, Rosenberg AJWP, Groll J, Bouten CVC, Kurniawan NA, Smits AIPM, Gawlitta D. Layer-specific cell differentiation in bi-layered vascular grafts under flow perfusion. Biofabrication 2019; 12:015009. [PMID: 31553965 DOI: 10.1088/1758-5090/ab47f0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bioengineered grafts have the potential to overcome the limitations of autologous and non-resorbable synthetic vessels as vascular substitutes. However, one of the challenges in creating these living grafts is to induce and maintain multiple cell phenotypes with a biomimetic organization. Our biomimetic grafts with heterotypic design hold promises for functional neovessel regeneration by guiding the layered cellular and tissue organization into a native-like structure. In this study, a perfusable two-compartment bioreactor chamber was designed for the further maturation of these vascular grafts, with a compartmentalized exposure of the graft's luminal and outer layer to cell-specific media. We used the system for a co-culture of endothelial colony forming cells and multipotent mesenchymal stromal cells (MSCs) in the vascular grafts, produced by combining electrospinning and melt electrowriting. It was demonstrated that the targeted cell phenotypes (i.e. endothelial cells (ECs) and vascular smooth muscle cells (vSMCs), respectively) could be induced and maintained during flow perfusion. The confluent luminal layer of ECs showed flow responsiveness, as indicated by the upregulation of COX-2, KLF2, and eNOS, as well as through stress fiber remodeling and cell elongation. In the outer layer, the circumferentially oriented, multi-layered structure of MSCs could be successfully differentiated into vSM-like cells using TGFβ, as indicated by the upregulation of αSMA, calponin, collagen IV, and (tropo)elastin, without affecting the endothelial monolayer. The cellular layers inhibited diffusion between the outer and the inner medium reservoirs. This implies tightly sealed cellular layers in the constructs, resulting in truly separated bioreactor compartments, ensuring the exposure of the inner endothelium and the outer smooth muscle-like layer to cell-specific media. In conclusion, using this system, we successfully induced layer-specific cell differentiation with a native-like cell organization. This co-culture system enables the creation of biomimetic neovessels, and as such can be exploited to investigate and improve bioengineered vascular grafts.
Collapse
Affiliation(s)
- Iris Pennings
- Department of Oral and Maxillofacial Surgery & Special Dental Care, UMC Utrecht, Utrecht University, Utrecht, The Netherlands. Regenerative Medicine Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Klotz BJ, Oosterhoff LA, Utomo L, Lim KS, Vallmajo-Martin Q, Clevers H, Woodfield TBF, Rosenberg AJWP, Malda J, Ehrbar M, Spee B, Gawlitta D. A Versatile Biosynthetic Hydrogel Platform for Engineering of Tissue Analogues. Adv Healthc Mater 2019; 8:e1900979. [PMID: 31402634 PMCID: PMC7116179 DOI: 10.1002/adhm.201900979] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Indexed: 01/14/2023]
Abstract
For creating functional tissue analogues in tissue engineering, stem cells require very specific 3D microenvironments to thrive and mature. Demanding (stem) cell types that are used nowadays can find such an environment in a heterogeneous protein mixture with the trade name Matrigel. Several variations of synthetic hydrogel platforms composed of poly(ethylene glycol) (PEG), which are spiked with peptides, have been recently developed and shown equivalence to Matrigel for stem cell differentiation. Here a clinically relevant hydrogel platform, based on PEG and gelatin, which even outperforms Matrigel when targeting 3D prevascularized bone and liver organoid tissue engineering models is presented. The hybrid hydrogel with natural and synthetic components stimulates efficient cell differentiation, superior to Matrigel models. Furthermore, the strength of this hydrogel lies in the option to covalently incorporate unmodified proteins. These results demonstrate how a hybrid hydrogel platform with intermediate biological complexity, when compared to existing biological materials and synthetic PEG-peptide approaches, can efficiently support tissue development from human primary cells.
Collapse
Affiliation(s)
- Barbara J. Klotz
- Department of Oral and Maxillofacial Surgery and Special Dental
Care, University Medical Center Utrecht, Utrecht University, 3508 GA
Utrecht, the Netherlands; Regenerative Medicine Utrecht, 3584 CT Utrecht,
the Netherlands
| | - Loes A. Oosterhoff
- Department of Clinical Sciences of Companion Animals, Faculty of
Veterinary Medicine, Utrecht University, 3508 TC Utrecht, the
Netherlands
| | - Lizette Utomo
- Department of Oral and Maxillofacial Surgery and Special Dental
Care, University Medical Center Utrecht, Utrecht University 3508 GA Utrecht,
the Netherlands; Regenerative Medicine Utrecht, 3584 CT Utrecht, the
Netherlands
| | - Khoon S. Lim
- Department of Orthopaedic Surgery and Musculoskeletal Medicine,
Centre for Bioengineering and Nanomedicine, University of Otago,
Christchurch 8011, New Zealand
| | - Queralt Vallmajo-Martin
- Department of Obstetrics, University Hospital Zurich, University of
Zurich, 8091 Zurich, Switzerland
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences,
University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - Tim B. F. Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal Medicine,
Centre for Bioengineering and Nanomedicine, University of Otago,
Christchurch 8011, New Zealand
| | - Antoine J. W. P. Rosenberg
- Department of Oral and Maxillofacial Surgery and Special Dental
Care, University Medical Center Utrecht, Utrecht University, 3508 GA
Utrecht, the Netherlands
| | - Jos Malda
- Regenerative Medicine Utrecht, 3584 CT Utrecht, the Netherlands;
Department of Orthopaedics, University Medical Center Utrecht, Utrecht
University, 3508 TC Utrecht, the Netherlands; Department of Equine Sciences,
Faculty of Veterinary Medicine, Utrecht University, 3508 TC Utrecht, the
Netherlands
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zurich, University
of Zurich, 8091 Zurich, Switzerland
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of
Veterinary Medicine, Utrecht University, 3508 TC Utrecht, the
Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery and Special Dental
Care, University Medical Center Utrecht, Utrecht University, 3508 GA
Utrecht, the Netherlands; Regenerative Medicine Utrecht, 3584 CT Utrecht,
the Netherlands
| |
Collapse
|
32
|
Bernal PN, Delrot P, Loterie D, Li Y, Malda J, Moser C, Levato R. Volumetric Bioprinting of Complex Living-Tissue Constructs within Seconds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1904209. [PMID: 31423698 DOI: 10.1002/adma.201904209] [Citation(s) in RCA: 226] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/29/2019] [Indexed: 05/17/2023]
Abstract
Biofabrication technologies, including stereolithography and extrusion-based printing, are revolutionizing the creation of complex engineered tissues. The current paradigm in bioprinting relies on the additive layer-by-layer deposition and assembly of repetitive building blocks, typically cell-laden hydrogel fibers or voxels, single cells, or cellular aggregates. The scalability of these additive manufacturing technologies is limited by their printing velocity, as lengthy biofabrication processes impair cell functionality. Overcoming such limitations, the volumetric bioprinting of clinically relevant sized, anatomically shaped constructs, in a time frame ranging from seconds to tens of seconds is described. An optical-tomography-inspired printing approach, based on visible light projection, is developed to generate cell-laden tissue constructs with high viability (>85%) from gelatin-based photoresponsive hydrogels. Free-form architectures, difficult to reproduce with conventional printing, are obtained, including anatomically correct trabecular bone models with embedded angiogenic sprouts and meniscal grafts. The latter undergoes maturation in vitro as the bioprinted chondroprogenitor cells synthesize neo-fibrocartilage matrix. Moreover, free-floating structures are generated, as demonstrated by printing functional hydrogel-based ball-and-cage fluidic valves. Volumetric bioprinting permits the creation of geometrically complex, centimeter-scale constructs at an unprecedented printing velocity, opening new avenues for upscaling the production of hydrogel-based constructs and for their application in tissue engineering, regenerative medicine, and soft robotics.
Collapse
Affiliation(s)
- Paulina Nuñez Bernal
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3584CX, Utrecht, The Netherlands
| | - Paul Delrot
- Laboratory of Applied Photonics Devices, École Polytechnique Fédéral Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Damien Loterie
- Laboratory of Applied Photonics Devices, École Polytechnique Fédéral Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Yang Li
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3584CX, Utrecht, The Netherlands
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3584CX, Utrecht, The Netherlands
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584CX, Utrecht, The Netherlands
| | - Christophe Moser
- Laboratory of Applied Photonics Devices, École Polytechnique Fédéral Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3584CX, Utrecht, The Netherlands
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584CX, Utrecht, The Netherlands
| |
Collapse
|
33
|
Cidonio G, Alcala-Orozco CR, Lim KS, Glinka M, Mutreja I, Kim YH, Dawson JI, Woodfield TBF, Oreffo ROC. Osteogenic and angiogenic tissue formation in high fidelity nanocomposite Laponite-gelatin bioinks. Biofabrication 2019; 11:035027. [PMID: 30991370 DOI: 10.1088/1758-5090/ab19fd] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
34
|
Hoffman T, Khademhosseini A, Langer R. Chasing the Paradigm: Clinical Translation of 25 Years of Tissue Engineering. Tissue Eng Part A 2019; 25:679-687. [PMID: 30727841 PMCID: PMC6533781 DOI: 10.1089/ten.tea.2019.0032] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 12/31/2022] Open
Abstract
IMPACT STATEMENT In this Perspective, we discuss the impact of the past 25 years of tissue engineering on the development of clinical therapies. Based on their success and other significant research accomplishments, platforms of innovation were identified. Their discoveries will enable tissue engineering inspired therapies to meet the requirements necessary for large-scale manufacturing and Food and Drug Administration (FDA) approval for a diverse range of indications.
Collapse
Affiliation(s)
- Tyler Hoffman
- Department of Bioengineering, University of California, Los Angeles, California
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California
- California NanoSystems Institute (CNSI), University of California, Los Angeles, California
| | - Ali Khademhosseini
- Department of Bioengineering, University of California, Los Angeles, California
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California
- California NanoSystems Institute (CNSI), University of California, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, California
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
35
|
Sarker MD, Naghieh S, Sharma NK, Ning L, Chen X. Bioprinting of Vascularized Tissue Scaffolds: Influence of Biopolymer, Cells, Growth Factors, and Gene Delivery. JOURNAL OF HEALTHCARE ENGINEERING 2019; 2019:9156921. [PMID: 31065331 PMCID: PMC6466897 DOI: 10.1155/2019/9156921] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/03/2019] [Indexed: 01/16/2023]
Abstract
Over the past decades, tissue regeneration with scaffolds has achieved significant progress that would eventually be able to solve the worldwide crisis of tissue and organ regeneration. While the recent advancement in additive manufacturing technique has facilitated the biofabrication of scaffolds mimicking the host tissue, thick tissue regeneration remains challenging to date due to the growing complexity of interconnected, stable, and functional vascular network within the scaffold. Since the biological performance of scaffolds affects the blood vessel regeneration process, perfect selection and manipulation of biological factors (i.e., biopolymers, cells, growth factors, and gene delivery) are required to grow capillary and macro blood vessels. Therefore, in this study, a brief review has been presented regarding the recent progress in vasculature formation using single, dual, or multiple biological factors. Besides, a number of ways have been presented to incorporate these factors into scaffolds. The merits and shortcomings associated with the application of each factor have been highlighted, and future research direction has been suggested.
Collapse
Affiliation(s)
- M. D. Sarker
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Saman Naghieh
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - N. K. Sharma
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Liqun Ning
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
36
|
Moses JC, Dey M, Devi KB, Roy M, Nandi SK, Mandal BB. Synergistic Effects of Silicon/Zinc Doped Brushite and Silk Scaffolding in Augmenting the Osteogenic and Angiogenic Potential of Composite Biomimetic Bone Grafts. ACS Biomater Sci Eng 2019; 5:1462-1475. [PMID: 33405621 DOI: 10.1021/acsbiomaterials.8b01350] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cell instructive scaffolding platforms displaying synergistic effects by virtue of their chemical and physical cues have tremendous scope in modulating cell phenotype and thus improving the success of any graft. In this regard, we report here the development of Si- and Zn-doped brushite cement composited with silk scaffolding that hierarchically emulated the cancellous bone. The composite scaffolds fabricated exhibited an open porous network capable of enhanced osteoblast survival as attested by increased alkaline phosphatase activity and also sustaining osteoclast activity affirmed by tartrate resistant acid phosphatase staining. Moreover, the chemical cues presented by dissolutions products from the composite scaffold enabled the osteoblasts to secrete proangiogenic factors which favored better endothelial cell survival, confirmed through in vitro experiments. Moreover, the efficacy of these composite biomimetic scaffolds was validated in vivo in volumetric femur defects in rabbits, which revealed that these matrices influenced vascular cell infiltration and favored the formation of matured bony plate. Fluorochrome labeling studies and microtomography analysis revealed that at the end of three months, the implanted composite scaffolds had completely resorbed, leaving behind neo-osseous tissue and vouching for clinical translation of these composite matrices as viable and affordable bone-graft substitutes.
Collapse
Affiliation(s)
- Joseph Christakiran Moses
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Mainak Dey
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata 700037, West Bengal, India
| | - K Bavya Devi
- Department of Metallurgical and Materials Engineering, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Mangal Roy
- Department of Metallurgical and Materials Engineering, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata 700037, West Bengal, India
| | - Biman B Mandal
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| |
Collapse
|