1
|
Ferguson BM, Clark JR, Li Q. Scaffold geometries designed to promote bone ingrowth by enhancing mechanobiological stimulation and biotransportation - A multiobjective optimisation approach. J Mech Behav Biomed Mater 2025; 164:106883. [PMID: 39919445 DOI: 10.1016/j.jmbbm.2024.106883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/20/2024] [Accepted: 12/26/2024] [Indexed: 02/09/2025]
Abstract
In a tissue-engineered bone scaffold implant, the process of neo-tissue ingrowth and remodelling into hard lamellar bone occurs slowly; it typically requires a period of several months to a year (or more) to complete. This research considers the design optimisation of a scaffold's unit cell geometry for the purpose of accelerating the rate at which neo-tissue forms in the porous network of the scaffold (ingrowth), and hence, reduce the length of time to complete the bone ingrowth process. In this study, the basic structure of the scaffold is the Schwarz Primitive (P) surface unit cell, selected for its compelling biomechanical and permeability characteristics. The geometry of the scaffold is varied using two parameters (namely iso-value, k, and spatial period, a) within the surface equation defining the Schwarz P-surface unit cell. In total, sixteen different unit cell geometries are considered here with the porosity ranging from 50% to 82%. The design objectives for the scaffold are to (i) enhance mechanobiological stimulus conditions conducive to bone apposition and (ii) enhance permeability to improve the transport of nutrients/oxygen and metabolities to and from the sites of neo-tissue formation throughout the porous scaffold. The independent design variables (k and a) of the periodic unit cell geometry are optimised to best satisfy the design objectives of appositional mechanobiological stimulus and biotransporting permeability. First, a reconstructed sheep mandible computed tomographic (CT)-based finite element (FE) analysis model is used to relate the strain energy density and mechanobiological stimulus to the design variables. Next, a computational fluid dynamics (CFD) model of a 5 × 5 × 5 unit cell scaffold is developed to relate the distributions of pressure and fluid velocity to the design variables. Then, surrogate modelling is undertaken in which bivariate cubic polynomial response surfaces are fitted to the FE and CFD analysis output data to form mathematical functions of each objective with respect to the two design variables. Finally, a multiobjective optimisation algorithm is invoked to determine the best trade-off between the competing design objectives of mechanobiological stimulus and biofluidic permeability. The novel design of the scaffold structure is anticipated to provide a better biomechanical and biotransport environment for tissue regeneration.
Collapse
Affiliation(s)
- Ben M Ferguson
- School of Aerospace, Mechanical and Mechatronic Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia.
| | - Jonathan R Clark
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, 2050, Australia; Central Clinical School, Faculty of Medicine and Health, The University of Sydney, University of Sydney, NSW, 2006, Australia; . Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Sydney, NSW, 2050, Australia
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia
| |
Collapse
|
2
|
De Rosa CA, Wright CJ, Xiong Y, Del Giudice F, Zhao F. Graded porous scaffold mediates internal fluidic environment for 3D in vitro mechanobiology. Comput Biol Med 2025; 186:109674. [PMID: 39809085 DOI: 10.1016/j.compbiomed.2025.109674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 12/13/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Most cell types are mechanosensitive, their activities such as differentiation, proliferation and apoptosis, can be influenced by the mechanical environment through mechanical stimulation. In three dimensional (3D) mechanobiological in vitro studies, the porous structure of scaffold controls the local mechanical environment that applied to cells. Many previous studies have focused on the topological design of homogeneous scaffold struts. However, the impact of scaffold inhomogeneity on the mechanical environment, which is essential in mechanobiological application (e.g. for multi-cells co-culture), remains elusive. In this study, we use a computational fluid dynamics (CFD) approach together with data analysis to study the influence of a porosity gradient (10 %-30 % porosity difference) on the local and global mechanical environment (wall shear stress - WSS) within the commonly used structures of triple periodic minimal surfaces (TPMS). In addition, the anisotropy of internal WSS and scaffold permeability caused by the porosity gradient is investigated. It is found that the influence of anisotropy on the average WSS and permeability is up to 11 % and 31 %, respectively. These results, as theoretical references will be useful to tissue engineers and mechanobiologists for scaffold design and in vitro experiment planning such as integrated use of graded scaffold and bioreactors for specific cell types.
Collapse
Affiliation(s)
- Chiara Angela De Rosa
- Department of Biomedical Engineering, Faculty of Science and Engineering, Swansea University, Swansea, United Kingdom; Zienkiewicz Institute for Modelling Data and AI, Swansea University, Swansea, United Kingdom
| | - Christopher J Wright
- Department of Biomedical Engineering, Faculty of Science and Engineering, Swansea University, Swansea, United Kingdom
| | - Yi Xiong
- School of System Design and Intelligent Manufacturing, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Francesco Del Giudice
- Complex Fluid Research Group, Department of Chemical Engineering, Faculty of Science and Engineering, Swansea University, Swansea, United Kingdom
| | - Feihu Zhao
- Department of Biomedical Engineering, Faculty of Science and Engineering, Swansea University, Swansea, United Kingdom; Zienkiewicz Institute for Modelling Data and AI, Swansea University, Swansea, United Kingdom.
| |
Collapse
|
3
|
Gadgil V, Kumbhojkar S, Sapre T, Deshmukh P, Dhatrak P. Investigating wall shear stress and the static pressure in bone scaffolds: a study of porosity and fluid flow dynamics. Biomech Model Mechanobiol 2025; 24:185-195. [PMID: 39477830 DOI: 10.1007/s10237-024-01904-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 10/19/2024] [Indexed: 02/24/2025]
Abstract
In bone tissue engineering, scaffolds are crucial as they provide a suitable structure for cell proliferation. Transporting Dulbecco's Modified Eagle Medium (DMEM) to the cells and regulating the scaffold's biocompatibility are both controlled by the dynamics of the fluid passing through the scaffold pores. Scaffold design selection and modeling are thus important in tissue engineering to achieve successful bone regeneration. This study aims to design and analyze three scaffold designs-Face-Centered Cubic (FCC), and two newly developed designs Octagonal Truss and a Square Pyramid with four porosity variations. The research aims to analyze the effect of design and porosity variation on pressure and wall shear stress, essential for analyzing scaffold biocompatibility in tissue engineering. Three scaffold designs with varying porosities with strut diameters ranging from 0.3 to 0.6 mm were modeled to analyze the behavior using BioMed Clear Resin. The fluid dynamics within these scaffolds were then examined using Computational Fluid Dynamics (CFD) to understand how different porosity levels affect fluid flow pressure and wall shear stress. The findings revealed variations in wall shear stress and their influence on cell proliferation. The maximum value of wall shear stress (WSS) is observed in the Square Pyramid model. The analysis shows that WSS at the inlet decreases as strut diameters increase or porosity percentages rise offering valuable insights for the development of effective scaffold designs. It can be concluded from the results that the Square Pyramid design has the highest value of WSS, thus increasing the chances of cell growth. From a biological perspective, the results of this work show promise for creating better scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Vedang Gadgil
- Department of Mechanical Engineering, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, India.
| | - Shriram Kumbhojkar
- Department of Mechanical Engineering, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, India
| | - Tushar Sapre
- Department of Mechanical Engineering, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, India
| | - Prathamesh Deshmukh
- Department of Mechanical Engineering, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, India
| | - Pankaj Dhatrak
- Department of Mechanical Engineering, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, India
| |
Collapse
|
4
|
Josephson TO, Morgan EF. Mechanobiological optimization of scaffolds for bone tissue engineering. Biomech Model Mechanobiol 2024; 23:2025-2042. [PMID: 39060881 DOI: 10.1007/s10237-024-01880-0] [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: 03/18/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Synthetic bone graft scaffolds aim to generate new bone tissue and alleviate the limitations of autografts and allografts. To meet that aim, it is essential to have a design approach able to generate scaffold architectures that will promote bone formation. Here, we present a topology-varying design optimization method, the "mixed-topology" approach, that generates new designs from a set of starting structures. This approach was used with objective functions focusing on improving the scaffold's local mechanical microenvironments to mechanobiologically promote bone formation within the scaffold and constraints to ensure manufacturability and achieve desired macroscale properties. The results demonstrate that this approach can successfully generate scaffold designs with improved microenvironments, taking into account different combinations of relevant stimuli and constraints.
Collapse
Affiliation(s)
- Timothy O Josephson
- Biomedical Engineering, Boston University, Boston, MA, USA.
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, USA.
| | - Elise F Morgan
- Biomedical Engineering, Boston University, Boston, MA, USA
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, USA
- Mechanical Engineering, Boston University, Boston, MA, USA
| |
Collapse
|
5
|
Bell JA, Mayfield CK, Collon K, Chang S, Gallo MC, Lechtholz-Zey E, Ayad M, Sugiyam O, Tang AH, Park SH, Lieberman JR. In vivo effects of cell seeding technique in an ex vivo regional gene therapy model for bone regeneration. J Biomed Mater Res A 2024; 112:1688-1698. [PMID: 38602243 DOI: 10.1002/jbm.a.37718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/14/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024]
Abstract
When delivering cells on a scaffold to treat a bone defect, the cell seeding technique determines the number and distribution of cells within a scaffold, however the optimal technique has not been established. This study investigated if human adipose-derived stem cells (ASCs) transduced with a lentiviral vector to overexpress bone morphogenetic protein 2 (BMP-2) and loaded on a scaffold using dynamic orbital shaker could reduce the total cell dose required to heal a critical sized bone defect when compared with static seeding. Human ASCs were loaded onto a collagen/biphasic ceramic scaffold using static loading and dynamic orbital shaker techniques, compared with our labs standard loading technique, and implanted into femoral defects of nude rats. Both a low dose and standard dose of transduced cells were evaluated. Outcomes investigated included BMP-2 production, radiographic healing, micro-computerized tomography, histologic assessment, and biomechanical torsional testing. BMP-2 production was higher in the orbital shaker cohort compared with the static seeding cohort. No statistically significant differences were noted in radiographic, histomorphometric, and biomechanical outcomes between the low-dose static and dynamic seeding groups, however the standard-dose static seeding cohort had superior biomechanical properties. The standard-dose 5 million cell dose standard loading cohort had superior maximum torque and torsional stiffness on biomechanical testing. The use of orbital shaker technique was labor intensive and did not provide equivalent biomechanical results with the use of fewer cells.
Collapse
Affiliation(s)
- Jennifer A Bell
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Cory K Mayfield
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Kevin Collon
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Stephanie Chang
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Matthew C Gallo
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Elizabeth Lechtholz-Zey
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Mina Ayad
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Osamu Sugiyam
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Amy H Tang
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Sang-Hyun Park
- J. Vernon Luck Orthopaedic Research Center, Orthopaedic Institute for Children, Los Angeles, California, USA
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| |
Collapse
|
6
|
Lin CL, Su YW, Chen YW, Kuo CH, Tu TY, Tsai JC, Shyong YJ. BMSC loaded photo-crosslinked hyaluronic acid/collagen hydrogel incorporating FG4592 for enhanced cell proliferation and nucleus pulposus differentiation. Int J Biol Macromol 2024; 273:132828. [PMID: 38834125 DOI: 10.1016/j.ijbiomac.2024.132828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 05/10/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
Intervertebral disc degeneration arises from damage or degeneration of the nucleus pulposus (NP). In this study, we developed a photo-crosslinkable hydrogel incorporating FG4592 to support the growth and differentiation of bone-marrow-derived mesenchymal stem cells (BMSC). Initially, hyaluronic acid was modified with tyramine and combined with collagen to introduce riboflavin as a photo-crosslinker. This hydrogel transitioned from liquid to gel upon exposure to blue light in 3 min. The results showed that the hydrogel was biodegradable and had mechanical properties comparable to those of human NP tissues. Scanning electron microscopy after BMSC seeding in the hydrogel revealed an even distribution, and cells adhered to the collagen fibers in the hydrogel with minimal cell mortality. The effect of FG4592 on BMSC proliferation and differentiation was examined, revealing the capability of FG4592 to promote BMSC proliferation and direct differentiation resembling human NP cells. After cultivating BMSCs in the photo-crosslinked hydrogel, there was an upregulation in the expression of glycosaminoglycans, aggrecan, type II collagen, and keratin 19 proteins. Cross-species analyses of rat and human BMSCs revealed consistent results. For potential clinical applications, BMSC loaded with photo-crosslinked hydrogels can be injected into damaged intervertebral disc to facilitate NP regeneration.
Collapse
Affiliation(s)
- Cheng-Li Lin
- Department of Orthopedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Wen Su
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Wei Chen
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Hsiang Kuo
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Jui-Chen Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yan-Jye Shyong
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
7
|
Zhang B. Computational fluid dynamics analysis of the fluid environment of 3D printed gradient structure in interfacial tissue engineering. Med Eng Phys 2024; 128:104173. [PMID: 38789213 DOI: 10.1016/j.medengphy.2024.104173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/19/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024]
Abstract
Mass transport properties within three-dimensional (3D) scaffold are essential for tissue regeneration, such as various fluid environmental cues influence mesenchymal stem cells differentiation. Recently, 3D printing has been emerging as a new technology for scaffold fabrication by controlling the scaffold pore geometry to affect cell growth environment. In this study, the flow field within scaffolds in a perfusion system was investigated with uniform structures, single gradient structures and complex gradient structures using computational fluid dynamics (CFD) method. The CFD results from those uniform structures indicate the fluid velocity and fluid shear stress within the scaffold structure increased as the filament diameter increasing, pore width decreasing, pore shape decreased from 90° to 15°, and layer configuration changing from lattice to stagger structure. By assembling those uniform structure as single gradient structures, it is noted that the fluid dynamic characterisation within the scaffold remains the same as the corresponding uniform structures. A complex gradient structure was designed to mimic natural osteochondral tissue by assembly the uniform structures of filament diameter, pore width, pore shape and layer configuration. The results show that the fluid velocity and fluid shear stress within the complex gradient structure distribute gradually increasing and their maximum magnitude were from 1.15 to 3.20 mm/s, and from 12 to 39 mPa, respectively. CFD technique allows the prediction of velocity and fluid shear stress within the designed 3D gradient scaffolds, which would be beneficial for the tissue scaffold development for interfacial tissue engineering in the future.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Mechanical and Aerospace, Brunel University London, London, United Kingdom.
| |
Collapse
|
8
|
Seehanam S, Khrueaduangkham S, Sinthuvanich C, Sae-Ueng U, Srimaneepong V, Promoppatum P. Evaluating the effect of pore size for 3d-printed bone scaffolds. Heliyon 2024; 10:e26005. [PMID: 38375289 PMCID: PMC10875428 DOI: 10.1016/j.heliyon.2024.e26005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
The present study investigated the influence of pore size of strut-based Diamond and surface-based Gyroid structures for their suitability as medical implants. Samples were made additively from laser powder bed fusion process with a relative density of 0.3 and pore sizes ranging from 300 to 1300 μm. They were subsequently examined for their manufacturability and mechanical properties. In addition, non-Newtonian computational fluid dynamics and discrete phase models were conducted to assess pressure drop and cell seeding efficiency. The results showed that both Diamond and Gyroid had higher as-built densities with smaller pore sizes. However, Gyroid demonstrated better manufacturability as its relative density was closer to the as-designed one. In addition, based on mechanical testing, the elastic modulus was largely unaffected by pore size, but post-yielding behaviors differed, especially in Diamond. High mechanical sensitivity in Diamond could be explained partly by Finite Element simulations, which revealed stress localization in Diamond and more uniform stress distribution in Gyroid. Furthermore, we defined the product of the normalized specific surface, normalized pressure drop, and cell seeding efficiency as the indicator of an optimal pore size, in which this factor identified an optimal pore size of approximately 500 μm for both Diamond and Gyroid. Besides, based on such criterion, Gyroid exhibited greater applicability as bone scaffolds. In summary, this study provides comprehensive assessment of the effect of pore size and demonstrates the efficient estimation of an in-silico framework for evaluating lattice structures as medical implants, which could be applied to other lattice architectures.
Collapse
Affiliation(s)
- Saran Seehanam
- Center for Lightweight Materials, Design, and Manufacturing, Department of Mechanical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi (KMUTT), Bangmod, Bangkok, 10140, Thailand
| | - Suppakrit Khrueaduangkham
- Center for Lightweight Materials, Design, and Manufacturing, Department of Mechanical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi (KMUTT), Bangmod, Bangkok, 10140, Thailand
| | - Chomdao Sinthuvanich
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Udom Sae-Ueng
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Viritpon Srimaneepong
- Department of Prosthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Patcharapit Promoppatum
- Center for Lightweight Materials, Design, and Manufacturing, Department of Mechanical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi (KMUTT), Bangmod, Bangkok, 10140, Thailand
- OsseoLabs Co. Ltd., Bangkok, 10400, Thailand
| |
Collapse
|
9
|
Chmielewska A, Dean D. The role of stiffness-matching in avoiding stress shielding-induced bone loss and stress concentration-induced skeletal reconstruction device failure. Acta Biomater 2024; 173:51-65. [PMID: 37972883 DOI: 10.1016/j.actbio.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
It is well documented that overly stiff skeletal replacement and fixation devices may fail and require revision surgery. Recent attempts to better support healing and sustain healed bone have looked at stiffness-matching of these devices to the desired role of limiting the stress on fractured or engrafted bone to compressive loads and, after the reconstructed bone has healed, to ensure that reconstructive medical devices (implants) interrupt the normal loading pattern as little as possible. The mechanical performance of these devices can be optimized by adjusting their location, integration/fastening, material(s), geometry (external and internal), and surface properties. This review highlights recent research that focuses on the optimal design of skeletal reconstruction devices to perform during and after healing as the mechanical regime changes. Previous studies have considered auxetic materials, homogeneous or gradient (i.e., adaptive) porosity, surface modification to enhance device/bone integration, and choosing the device's attachment location to ensure good osseointegration and resilient load transduction. By combining some or all of these factors, device designers work hard to avoid problems brought about by unsustainable stress shielding or stress concentrations as a means of creating sustainable stress-strain relationships that best repair and sustain a surgically reconstructed skeletal site. STATEMENT OF SIGNIFICANCE: Although standard-of-care skeletal reconstruction devices will usually allow normal healing and improved comfort for the patient during normal activities, there may be significant disadvantages during long-term use. Stress shielding and stress concentration are amongst the most common causes of failure of a metallic device. This review highlights recent developments in devices for skeletal reconstruction that match the stiffness, while not interrupting the normal loading pattern of a healthy bone, and help to combat stress shielding and stress concentration. This review summarises various approaches to achieve stiffness-matching: application of materials with modulus close to that of the bone; adaptation of geometry with pre-defined mechanical properties; and/or surface modification that ensures good integration and proper load transfer to the bone.
Collapse
Affiliation(s)
- Agnieszka Chmielewska
- The Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA.
| | - David Dean
- The Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Plastic & Reconstructive Surgery, The Ohio State University, Columbus, OH 43212, USA
| |
Collapse
|
10
|
Ivanovski S, Breik O, Carluccio D, Alayan J, Staples R, Vaquette C. 3D printing for bone regeneration: challenges and opportunities for achieving predictability. Periodontol 2000 2023; 93:358-384. [PMID: 37823472 DOI: 10.1111/prd.12525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/18/2023] [Accepted: 08/26/2023] [Indexed: 10/13/2023]
Abstract
3D printing offers attractive opportunities for large-volume bone regeneration in the oro-dental and craniofacial regions. This is enabled by the development of CAD-CAM technologies that support the design and manufacturing of anatomically accurate meshes and scaffolds. This review describes the main 3D-printing technologies utilized for the fabrication of these patient-matched devices, and reports on their pre-clinical and clinical performance including the occurrence of complications for vertical bone augmentation and craniofacial applications. Furthermore, the regulatory pathway for approval of these devices is discussed, highlighting the main hurdles and obstacles. Finally, the review elaborates on a variety of strategies for increasing bone regeneration capacity and explores the future of 4D bioprinting and biodegradable metal 3D printing.
Collapse
Affiliation(s)
- Saso Ivanovski
- School of Dentistry, Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Queensland, Herston, Australia
| | - Omar Breik
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, Queensland, Australia
| | - Danilo Carluccio
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, Queensland, Australia
| | - Jamil Alayan
- School of Dentistry, Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Queensland, Herston, Australia
| | - Ruben Staples
- School of Dentistry, Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Queensland, Herston, Australia
| | - Cedryck Vaquette
- School of Dentistry, Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Queensland, Herston, Australia
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
Mohol SS, Kumar M, Sharma V. PLA-based nature-inspired architecture for bone scaffolds: A finite element analysis. Comput Biol Med 2023; 163:107163. [PMID: 37329619 DOI: 10.1016/j.compbiomed.2023.107163] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/04/2023] [Accepted: 06/07/2023] [Indexed: 06/19/2023]
Abstract
The implantation of bio-degradable scaffolds is considered as a promising approach to address the repair of bone defects. This article aims to develop a computational approach to study the mechanical behaviour, fluid dynamic, and degradation impact on polylactic acid scaffolds with nature-inspired design structures. Scaffold design is considered to be one of the main factors for the regulation of mechanical characteristics and fluid flow dynamics. In this article, five scaffolds with different nature-inspired architectures have been designed within a specific porosity range. Based on finite element analysis, their mechanical behaviour and computational fluid dynamic study are performed to evaluate the respective properties of different scaffolds. In addition, diffusion-governed degradation analysis of the scaffolds has been performed to compute the total time required for the scaffold to degrade within a given environment. Based on the mechanical behaviour, the Spider-web architecture scaffold was found to have the least deformation, and also the lowest value of equivalent stress and strain. The Nautilus Shell architecture scaffold had the highest value of equivalent stress and strain. The permeability of all the scaffolds was found to meet the requirement of the cancellous bone. All computational fluid dynamics (CFD) results of wall shear stress are in line with the requirement for cell differentiation. It was observed that the Spider-web architecture scaffold had undergone the slowest degradation, and the Giant Water Lily architecture scaffold experienced the fastest degradation.
Collapse
Affiliation(s)
- Shubham Shankar Mohol
- Additive and Subtractive Manufacturing Lab, Department of Mechanical and Industrial Engineering, IIT Roorkee, India
| | - Mohit Kumar
- Additive and Subtractive Manufacturing Lab, Department of Mechanical and Industrial Engineering, IIT Roorkee, India
| | - Varun Sharma
- Additive and Subtractive Manufacturing Lab, Department of Mechanical and Industrial Engineering, IIT Roorkee, India; Department of Mechanical and Industrial Engineering, IIT Roorkee, India.
| |
Collapse
|
12
|
Babakhanova G, Agrawal A, Arora D, Horenberg A, Budhathoki JB, Dunkers JP, Chalfoun J, Bajcsy P, Simon CG. Three-dimensional, label-free cell viability measurements in tissue engineering scaffolds using optical coherence tomography. J Biomed Mater Res A 2023; 111:1279-1291. [PMID: 36916776 DOI: 10.1002/jbm.a.37528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/31/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023]
Abstract
In the field of tissue engineering, 3D scaffolds and cells are often combined to yield constructs that are used as therapeutics to repair or restore tissue function in patients. Viable cells are often required to achieve the intended mechanism of action for the therapy, where the live cells may build new tissue or may release factors that induce tissue regeneration. Thus, there is a need to reliably measure cell viability in 3D scaffolds as a quality attribute of a tissue-engineered medical product. Here, we developed a noninvasive, label-free, 3D optical coherence tomography (OCT) method to rapidly (2.5 min) image large sample volumes (1 mm3 ) to assess cell viability and distribution within scaffolds. OCT imaging was assessed using a model scaffold-cell system consisting of a polysaccharide-based hydrogel seeded with human Jurkat cells. Four test systems were used: hydrogel seeded with live cells, hydrogel seeded with heat-shocked or fixed dead cells and hydrogel without any cells. Time series OCT images demonstrated changes in the time-dependent speckle patterns due to refractive index (RI) variations within live cells that were not observed for pure hydrogel samples or hydrogels with dead cells. The changes in speckle patterns were used to generate live-cell contrast by image subtraction. In this way, objects with large changes in RI were binned as live cells. Using this approach, on average, OCT imaging measurements counted 326 ± 52 live cells per 0.288 mm3 for hydrogels that were seeded with 288 live cells (as determined by the acridine orange-propidium iodide cell counting method prior to seeding cells in gels). Considering the substantial uncertainties in fabricating the scaffold-cell constructs, such as the error from pipetting and counting cells, a 13% difference in the live-cell count is reasonable. Additionally, the 3D distribution of live cells was mapped within a hydrogel scaffold to assess the uniformity of their distribution across the volume. Our results demonstrate a real-time, noninvasive method to rapidly assess the spatial distribution of live cells within a 3D scaffold that could be useful for assessing tissue-engineered medical products.
Collapse
Affiliation(s)
- Greta Babakhanova
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Anant Agrawal
- Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Deepika Arora
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Allison Horenberg
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Jagat B Budhathoki
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Joy P Dunkers
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Joe Chalfoun
- Software and Systems Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Peter Bajcsy
- Software and Systems Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Carl G Simon
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| |
Collapse
|
13
|
Lu Q, Diao J, Wang Y, Feng J, Zeng F, Yang Y, Kuang Y, Zhao N, Wang Y. 3D printed pore morphology mediates bone marrow stem cell behaviors via RhoA/ROCK2 signaling pathway for accelerating bone regeneration. Bioact Mater 2023; 26:413-424. [PMID: 36969106 PMCID: PMC10036893 DOI: 10.1016/j.bioactmat.2023.02.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 03/28/2023] Open
Abstract
Bone bionics and structural engineering have sparked a broad interest in optimizing artificial scaffolds for better bone regeneration. However, the mechanism behind scaffold pore morphology-regulated bone regeneration remains unclear, making the structure design of scaffolds for bone repair challenging. To address this issue, we have carefully assessed diverse cell behaviors of bone mesenchymal stem cells (BMSCs) on the β-tricalcium phosphate (β-TCP) scaffolds with three representative pore morphologies (i.e., cross column, diamond, and gyroid pore unit, respectively). Among the scaffolds, BMSCs on the β-TCP scaffold with diamond pore unit (designated as D-scaffold) demonstrated enhanced cytoskeletal forces, elongated nucleus, faster cell mobility, and better osteogenic differentiation potential (for example, the alkaline phosphatase expression level in D-scaffold were 1.5-2 times higher than other groups). RNA-sequencing analysis and signaling pathway intervention revealed that Ras homolog gene family A (RhoA)/Rho-associated kinase-2 (ROCK2) has in-depth participated in the pore morphology-mediated BMSCs behaviors, indicating an important role of mechanical signaling transduction in scaffold-cell interactions. Finally, femoral condyle defect repair results showed that D-scaffold could effectively promote endogenous bone regeneration, of which the osteogenesis rate was 1.2-1.8 times higher than the other groups. Overall, this work provides insights into pore morphology-mediated bone regeneration mechanisms for developing novel bioadaptive scaffold designs.
Collapse
Affiliation(s)
- Qiji Lu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
| | - Jingjing Diao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- Medical Devices Research & Testing Center of SCUT, Guangzhou, 510006, PR China
| | - Yingqu Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Jianlang Feng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
| | - Fansen Zeng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
| | - Yan Yang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
| | - Yudi Kuang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
- Guangdong Institute of Advanced Biomaterials and Medical Devices, Guangzhou, 510535, PR China
- Corresponding author. National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China.
| | - Naru Zhao
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
- Corresponding author. School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China.
| | - Yingjun Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
- Guangdong Institute of Advanced Biomaterials and Medical Devices, Guangzhou, 510535, PR China
- Corresponding author. School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China.
| |
Collapse
|
14
|
Ntousi O, Roumpi M, Siogkas P, Deligianni D, Fotiadis DI. Computational Fluid Dynamic Analysis of customised 3D-printed bone scaffolds with different architectures. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083223 DOI: 10.1109/embc40787.2023.10340034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Through the recent years, tissue engineering has been proven as a solid substitute of autografts in the stimulation of bone tissue regeneration, through the development of three dimensional (3D) porous matrices, commonly known as scaffolds. In this work, we analysed two scaffold structures with 500μm pore size, by performing computational fluid dynamics simulations, to compare permeability, Wall Shear Stress (WSS), velocity and pressure distributions. Taking into account those parameters the geometry named as "PCL-50" was the best to anticipate showing a superior performance in supporting cell growth due to the improved flow characteristics in the scaffold.Clinical Relevance- Bone defects that require invasive surgical treatment with high risks in terms of success and effectiveness. Bone tissue engineering (BTE) in combination with the use of computational fluid dynamics (CFD) analysis tools aim to assist in designing optimal scaffolds that better promote bone growth and repair. The fluid dynamic characteristics of a porous scaffold plays a vital role in cell viability and cell growth, affecting the osteogenic performance of the scaffold.
Collapse
|
15
|
Zhang Z, Zhu J, Liu Y, Shao J, Xie S. Effects of cell deformability and adhesion strength on dynamic cell seeding: Cell-scale investigation via mesoscopic modeling. J Biomech 2023; 153:111589. [PMID: 37137273 DOI: 10.1016/j.jbiomech.2023.111589] [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: 11/08/2022] [Revised: 03/26/2023] [Accepted: 04/11/2023] [Indexed: 05/05/2023]
Abstract
The flow of cell suspension through a porous scaffold is a common process in dynamic cell seeding, which determines the initial distribution of cells for constructing tissue-engineered grafts. Physical insights into the transport and adhesion behaviors of cells in this process are of great significance to the precise control of cell density and its distribution in the scaffold. Revealing of dynamic mechanisms underlying these cell behaviors through experiments is still difficult. The numerical approach therefore plays an important role in such studies. However, existing studies have mostly focused on external factors (e.g., flow conditions and scaffold architecture) but ignored the intrinsic biomechanical properties of cells as well as their associated effects. The present work utilized a well-established mesoscopic model to simulate the dynamic cell seeding within a porous scaffold, based on which a thorough investigation of the effects of cell deformability and cell-scaffold adhesion strength on the seeding process was carried out. The results show that the increase in either the stiffness or the bond strength of cells would augment the firm-adhesion rate and thus enhance seeding efficiency. In comparison to cell deformability, bond strength seems to play a more dominant role. Especially in the cases with weak bond strength, remarkable losses of seeding efficiency and distribution uniformity are observed. Noteworthily, it is found that both the firm-adhesion rate and the seeding efficiency are quantiatively related to the adhesion strength which is measured as the detachment force, suggesting a straightforward way to estimate the seeding outcome.
Collapse
Affiliation(s)
- Ziying Zhang
- College of Mechanical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Junwei Zhu
- College of Mechanical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Yangyang Liu
- Department of Mechanical Engineering, National University of Singapore, 10 Kent Ridge Crescent, Singapore 119260, Singapore
| | - Jiaru Shao
- College of Mechanical Engineering, Chongqing University of Technology, Chongqing 400054, PR China.
| | - Shuangyi Xie
- College of Mechanical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| |
Collapse
|
16
|
Pattnaik A, Sanket AS, Pradhan S, Sahoo R, Das S, Pany S, Douglas TEL, Dandela R, Liu Q, Rajadas J, Pati S, De Smedt SC, Braeckmans K, Samal SK. Designing of gradient scaffolds and their applications in tissue regeneration. Biomaterials 2023; 296:122078. [PMID: 36921442 DOI: 10.1016/j.biomaterials.2023.122078] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/19/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Gradient scaffolds are isotropic/anisotropic three-dimensional structures with gradual transitions in geometry, density, porosity, stiffness, etc., that mimic the biological extracellular matrix. The gradient structures in biological tissues play a major role in various functional and metabolic activities in the body. The designing of gradients in the scaffold can overcome the current challenges in the clinic compared to conventional scaffolds by exhibiting excellent penetration capacity for nutrients & cells, increased cellular adhesion, cell viability & differentiation, improved mechanical stability, and biocompatibility. In this review, the recent advancements in designing gradient scaffolds with desired biomimetic properties, and their implication in tissue regeneration applications have been briefly explained. Furthermore, the gradients in native tissues such as bone, cartilage, neuron, cardiovascular, skin and their specific utility in tissue regeneration have been discussed in detail. The insights from such advances using gradient-based scaffolds can widen the horizon for using gradient biomaterials in tissue regeneration applications.
Collapse
Affiliation(s)
- Ananya Pattnaik
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - A Swaroop Sanket
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Sanghamitra Pradhan
- Department of Chemistry, Institute of Technical Education and Research, Siksha 'O' Anusandhan University, Bhubaneswar, 751030, Odisha, India
| | - Rajashree Sahoo
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Sudiptee Das
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Swarnaprbha Pany
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Timothy E L Douglas
- Engineering Department, Lancaster University, Lancaster, United Kingdom; Materials Science Institute, Lancaster University, Lancaster, United Kingdom
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Bhubaneswar, Odisha, India
| | - Qiang Liu
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Stanford University School of Medicine, Department of Medicine, Stanford University, California, 94304, USA
| | - Jaykumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Stanford University School of Medicine, Department of Medicine, Stanford University, California, 94304, USA; Department of Bioengineering and Therapeutic Sciences, University of California San Francusco (UCSF) School of Parmacy, California, USA
| | - Sanghamitra Pati
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, University of Ghent, Ghent, 9000, Belgium.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, University of Ghent, Ghent, 9000, Belgium
| | - Sangram Keshari Samal
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India.
| |
Collapse
|
17
|
Brodeur A, Winter A, Roy V, Touzel Deschênes L, Gros-Louis F, Ruel J. Spherical rotary cell seeding system for production of small-caliber tissue-engineered blood vessels with complex geometry. Sci Rep 2023; 13:3001. [PMID: 36810756 PMCID: PMC9944280 DOI: 10.1038/s41598-023-29825-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/10/2023] [Indexed: 02/23/2023] Open
Abstract
Entirely biological human tissue-engineered blood vessels (TEBV) were previously developed for clinical use. Tissue-engineered models have also proven to be valuable tools in disease modelling. Moreover, there is a need for complex geometry TEBV for study of multifactorial vascular pathologies, such as intracranial aneurysms. The main goal of the work reported in this article was to produce an entirely human branched small-caliber TEBV. The use of a novel spherical rotary cell seeding system allows effective and uniform dynamic cell seeding for a viable in vitro tissue-engineered model. In this report, the design and fabrication of an innovative seeding system with random spherical 360° rotation is described. Custom made seeding chambers are placed inside the system and hold Y-shaped polyethylene terephthalate glycol (PETG) scaffolds. The seeding conditions, such as cell concentration, seeding speed and incubation time were optimized via count of cells adhered on the PETG scaffolds. This spheric seeding method was compared to other approaches, such as dynamic and static seeding, and clearly shows uniform cell distribution on PETG scaffolds. With this simple to use spherical system, fully biological branched TEBV constructs were also produced by seeding human fibroblasts directly on custom-made complex geometry PETG mandrels. The production of patient-derived small-caliber TEBVs with complex geometry and optimized cellular distribution all along the vascular reconstructed may be an innovative way to model various vascular diseases such as intracranial aneurysms.
Collapse
Affiliation(s)
- Alyssa Brodeur
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Quebec City, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, QC Canada
| | - Alexandre Winter
- grid.23856.3a0000 0004 1936 8390Department of Mechanical Engineering, Faculty of Sciences and Engineering, Laval University, Quebec City, QC Canada
| | - Vincent Roy
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Quebec City, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, QC Canada
| | - Lydia Touzel Deschênes
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Quebec City, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, QC Canada
| | - François Gros-Louis
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Quebec City, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, QC Canada
| | - Jean Ruel
- Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, QC, Canada. .,Department of Mechanical Engineering, Faculty of Sciences and Engineering, Laval University, Quebec City, QC, Canada.
| |
Collapse
|
18
|
Pires THV, Dunlop JWC, Castro APG, Fernandes PR. Wall Shear Stress Analysis and Optimization in Tissue Engineering TPMS Scaffolds. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7375. [PMID: 36295440 PMCID: PMC9612273 DOI: 10.3390/ma15207375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/28/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023]
Abstract
When designing scaffolds for bone tissue engineering (BTE), the wall shear stress (WSS), due to the fluid flow inside the scaffold, is an important factor to consider as it influences the cellular process involved in new tissue formation. The present work analyzed the average WSS in Schwartz diamond (SD) and gyroid (SG) scaffolds with different surface topologies and mesh elements using computational fluid dynamics (CFD) analysis. It was found that scaffold meshes with a smooth surface topology with tetrahedral elements had WSS levels 35% higher than the equivalent scaffold with a non-smooth surface topology with hexahedral elements. The present work also investigated the possibility of implementing the optimization algorithm simulated annealing to aid in the design of BTE scaffolds with a specific average WSS, with the outputs showing that the algorithm was able to reach WSS levels in the vicinity of 5 mPa (physiological range) within the established limit of 100 iterations. This proved the efficacy of combining CFD and optimization methods in the design of BTE scaffolds.
Collapse
Affiliation(s)
- Tiago H. V. Pires
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - John W. C. Dunlop
- MorphoPhysics Group, Department of the Chemistry and Physics of Materials, University of Salzburg, 5020 Salzburg, Austria
| | - André P. G. Castro
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- ESTSetúbal, Instituto Politécnico de Setúbal, 2914-761 Setúbal, Portugal
| | - Paulo R. Fernandes
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| |
Collapse
|
19
|
Novel structural designs of 3D-printed osteogenic graft for rapid angiogenesis. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00212-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Conrad B, Yang F. Hydroxyapatite-coated gelatin microribbon scaffolds induce rapid endogenous cranial bone regeneration in vivo. BIOMATERIALS ADVANCES 2022; 140:213050. [PMID: 35917686 DOI: 10.1016/j.bioadv.2022.213050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/25/2022] [Accepted: 07/24/2022] [Indexed: 06/15/2023]
Abstract
Hydroxyapatite (HA) has a composition similar to mineral bone and has been used for coating macroporous scaffolds to enhance bone formation. However, previous macroporous scaffolds did not support minimally invasive delivery. Our lab has reported on gelatin-based microribbon (μRB) shaped hydrogels, which combine injectability with macroporosity and support cranial bone formation in an immunocompromised mouse model. However, gelatin alone was not sufficient to support cranial bone formation in immunocompetent animals. To overcome this challenge, here we evaluated two methods to incorporate HA into gelatin μRB scaffolds using either modified simulated body fluid (mSBF) or commercially available HA nanoparticles (HAnp). HA incorporation and distribution were characterized using scanning electron microscopy and energy-dispersive X-ray spectroscopy. While both methods enhanced MSC osteogenesis and mineralization, the mSBF method led to undesirable reduction in mechanical properties. HAnp-coated μRB scaffolds were further evaluated in an immunocompetent mouse cranial defect model. Acellular HAnp-coated gelatin μRB scaffolds induced rapid and robust endogenous cranial bone regeneration as shown by MicroCT imaging and histology. Co-delivery with exogenous MSCs led to later bone resorption accompanied by increased osteoclast activity. In summary, our results demonstrate the promise of gelatin μRBs with HAnps as a promising therapy for cranial bone regeneration without the need for exogenous cells or growth factors.
Collapse
Affiliation(s)
- Bogdan Conrad
- Program of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 240 Pasteur Dr., Biomedical Innovation Building 1200, Palo Alto, CA 94304, United States of America.
| | - Fan Yang
- Departments of Orthopaedic Surgery and Bioengineering Stanford University, 240 Pasteur Dr., Biomedical Innovation Building 1200, Palo Alto, CA 94304, United States of America.
| |
Collapse
|
21
|
Sarviya N, Basu SM, Mani R, Chauhan M, Kingshott P, Giri J. Biomimicking nanofibrous gelatin microspheres recreating the stem cell niche for their ex-vivo expansion and in-vivo like differentiation for injectable stem cell transplantation. BIOMATERIALS ADVANCES 2022; 139:212981. [PMID: 35882137 DOI: 10.1016/j.bioadv.2022.212981] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
Stem cells based novel treatment modality for degenerative and immune dysfunction diseases created a huge demand of suitable carriers to support ex-vivo production of quality stem cells, and effective in-vivo transplantation of stem cells and their fate. In spite of promising candidature of nanofibrous microspheres (NFM) to recreate native stem cell niches to be used for possible scaling-up for ex-vivo stem cells expansion, it remains fairly unexplored. A systematic study on the stem cell-NFM interaction comparative with commercial microspheres (CM) has been performed for the first time. Gelatin NFM with variable physicochemical properties such as size, surface properties, surface chemistry, and variable degradability were prepared using microemulsion coupled with thermally induced phase separation (TIPS) method. Effect of physicochemical properties of NFM and their cellular interaction such as binding, morphology, metabolic activity and proliferation studies were performed using human bone marrow-derived mesenchymal stem cells (hBMSCs), human dental follicle stem cells (hDFSCs) and human gingival fibroblast (HGF) cells and compared with the commercial and solid microspheres. Gelatin NFM supports excellent cell binding, proliferation, metabolic activities and chemical cues specific differentiation. All out-turns indicate that NFM stand to be an outstanding candidate for ex-vivo cells' expansion and injectable carriers for stem cell transplantation.
Collapse
Affiliation(s)
- Nandini Sarviya
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India; Department of Chemistry and Biotechnology, Swinburne Institute of Technology, Victoria, Australia
| | - Suparna Mercy Basu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Rajesh Mani
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Meenakshi Chauhan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Peter Kingshott
- Department of Chemistry and Biotechnology, Swinburne Institute of Technology, Victoria, Australia
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India.
| |
Collapse
|
22
|
Karaman D, Ghahramanzadeh Asl H. Biomechanical behavior of diamond lattice scaffolds obtained by two different design approaches with similar porosity; a numerical investigation with FEM and CFD analysis. Proc Inst Mech Eng H 2022; 236:794-810. [DOI: 10.1177/09544119221091346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Scaffolds provide a suitable environment for the bone tissue to maintain its self-healing ability and help new bone-cell formation by creating structures with similar mechanical properties to the surrounding tissue. In the modeling of the scaffolds, an optimum environment is tried to be provided by changing the geometrical properties of the cell architecture such as porosity, pore size, and specific surface area. For this purpose, different design approaches have been used in studies to change these properties. This study aims to determine whether scaffolds with similar porosities modeled by different design approaches exhibit distinct biomechanical behaviors or not. By using the Diamond lattice architecture, two different design approaches were constituted. The first approach has constant wall thickness and variable cell size, whereas the second approach contains variable wall thickness and constant cell size. The usage of different design approaches affected the amount of specific surface area in models with similar porosity. Mechanical compression tests were conducted via finite element analysis, while the permeability performance of configurations with similar porosities (50%, 60%, 70%, 80%, and 90%) was evaluated by using computational fluid dynamics. The mechanical results revealed that the structural strength decreased with increasing porosity. Since their higher specific surface area causes lower pressure drops, the second group exhibits better permeability. In addition, it was found that to evaluate the wall shear stresses occurring on the scaffold surfaces properly, it is essential to consider the stress distributions within the scaffold rather than the maximum values.
Collapse
Affiliation(s)
- Derya Karaman
- Department of Mechanical Engineering, Engineering Faculty, Karadeniz Technical University, Trabzon, Turkey
| | - Hojjat Ghahramanzadeh Asl
- Department of Mechanical Engineering, Engineering Faculty, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
23
|
Chan JK, Chadwick EA, Taniguchi D, Ahmadipour M, Suzuki T, Romero D, Amon C, Waddell TK, Karoubi G, Bazylak A. Cell Inertia: Predicting Cell Distributions in Lung Vasculature to Optimize Re-endothelialization. Front Bioeng Biotechnol 2022; 10:891407. [PMID: 35573256 PMCID: PMC9092599 DOI: 10.3389/fbioe.2022.891407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/11/2022] [Indexed: 12/05/2022] Open
Abstract
We created a transient computational fluid dynamics model featuring a particle deposition probability function that incorporates inertia to quantify the transport and deposition of cells in mouse lung vasculature for the re-endothelialization of the acellular organ. Our novel inertial algorithm demonstrated a 73% reduction in cell seeding efficiency error compared to two established particle deposition algorithms when validated with experiments based on common clinical practices. We enhanced the uniformity of cell distributions in the lung vasculature by increasing the injection flow rate from 3.81 ml/min to 9.40 ml/min. As a result, the cell seeding efficiency increased in both the numerical and experimental results by 42 and 66%, respectively.
Collapse
Affiliation(s)
- Jason K.D. Chan
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Eric A. Chadwick
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Daisuke Taniguchi
- Latner Thoracic Surgery Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| | - Mohammadali Ahmadipour
- Latner Thoracic Surgery Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering (BME), University of Toronto, Toronto, ON, Canada
| | - Takaya Suzuki
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - David Romero
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Cristina Amon
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering (BME), University of Toronto, Toronto, ON, Canada
| | - Thomas K. Waddell
- Latner Thoracic Surgery Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering (BME), University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Golnaz Karoubi
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
- Latner Thoracic Surgery Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Aimy Bazylak
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
- *Correspondence: Aimy Bazylak,
| |
Collapse
|
24
|
Bernal PN, Bouwmeester M, Madrid-Wolff J, Falandt M, Florczak S, Rodriguez NG, Li Y, Größbacher G, Samsom RA, van Wolferen M, van der Laan LJW, Delrot P, Loterie D, Malda J, Moser C, Spee B, Levato R. Volumetric Bioprinting of Organoids and Optically Tuned Hydrogels to Build Liver-Like Metabolic Biofactories. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110054. [PMID: 35166410 DOI: 10.1002/adma.202110054] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Organ- and tissue-level biological functions are intimately linked to microscale cell-cell interactions and to the overarching tissue architecture. Together, biofabrication and organoid technologies offer the unique potential to engineer multi-scale living constructs, with cellular microenvironments formed by stem cell self-assembled structures embedded in customizable bioprinted geometries. This study introduces the volumetric bioprinting of complex organoid-laden constructs, which capture key functions of the human liver. Volumetric bioprinting via optical tomography shapes organoid-laden gelatin hydrogels into complex centimeter-scale 3D structures in under 20 s. Optically tuned bioresins enable refractive index matching of specific intracellular structures, countering the disruptive impact of cell-mediated light scattering on printing resolution. This layerless, nozzle-free technique poses no harmful mechanical stresses on organoids, resulting in superior viability and morphology preservation post-printing. Bioprinted organoids undergo hepatocytic differentiation showing albumin synthesis, liver-specific enzyme activity, and remarkably acquired native-like polarization. Organoids embedded within low stiffness gelatins (<2 kPa) are bioprinted into mathematically defined lattices with varying degrees of pore network tortuosity, and cultured under perfusion. These structures act as metabolic biofactories in which liver-specific ammonia detoxification can be enhanced by the architectural profile of the constructs. This technology opens up new possibilities for regenerative medicine and personalized drug testing.
Collapse
Affiliation(s)
- Paulina Nuñez Bernal
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Manon Bouwmeester
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Jorge Madrid-Wolff
- Laboratory of Applied Photonics Devices, École Polytechnique Fédéral Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Marc Falandt
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Sammy Florczak
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Nuria Ginés Rodriguez
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Yang Li
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Gabriel Größbacher
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Roos-Anne Samsom
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Monique van Wolferen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Paul Delrot
- Readily3D SA, EPFL Innovation Park, Building A, Lausanne, CH-1015, Switzerland
| | - Damien Loterie
- Readily3D SA, EPFL Innovation Park, Building A, Lausanne, CH-1015, Switzerland
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Christophe Moser
- Laboratory of Applied Photonics Devices, École Polytechnique Fédéral Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| |
Collapse
|
25
|
Pires T, Dunlop JWC, Fernandes PR, Castro APG. Challenges in computational fluid dynamics applications for bone tissue engineering. Proc Math Phys Eng Sci 2022; 478:20210607. [PMID: 35153613 PMCID: PMC8791047 DOI: 10.1098/rspa.2021.0607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022] Open
Abstract
Bone injuries or defects that require invasive surgical treatment are a serious clinical issue, particularly when it comes to treatment success and effectiveness. Accordingly, bone tissue engineering (BTE) has been researching the use of computational fluid dynamics (CFD) analysis tools to assist in designing optimal scaffolds that better promote bone growth and repair. This paper aims to offer a comprehensive review of recent studies that use CFD analysis in BTE. The mechanical and fluidic properties of a given scaffold are coupled to each other via the scaffold architecture, meaning an optimization of one may negatively affect the other. For example, designs that improve scaffold permeability normally result in a decreased average wall shear stress. Linked with these findings, it appears there are very few studies in this area that state a specific application for their scaffolds and those that do are focused on in vitro bioreactor environments. Finally, this review also demonstrates a scarcity of studies that combine CFD with optimization methods to improve scaffold design. This highlights an important direction of research for the development of the next generation of BTE scaffolds.
Collapse
Affiliation(s)
- Tiago Pires
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - John W C Dunlop
- MorphoPhysics Group, Department of the Chemistry and Physics of Materials, University of Salzburg, Salzburg, Austria
| | | | - André P G Castro
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
26
|
Guarnera D, Iberite F, Piazzoni M, Gerges I, Santaniello T, Vannozzi L, Lenardi C, Ricotti L. Effects of the 3D Geometry Reconstruction on the Estimation of 3D Porous Scaffold Permeability . ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:4403-4407. [PMID: 34892196 DOI: 10.1109/embc46164.2021.9629664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
3D scaffolds for tissue engineering typically need to adopt a dynamic culture to foster cell distribution and survival throughout the scaffold. It is, therefore, crucial to know fluids' behavior inside the scaffold architecture, especially for complex porous ones. Here we report a comparison between simulated and measured permeability of a porous 3D scaffold, focusing on different modeling parameters. The scaffold features were extracted by microcomputed tomography (µCT) and representative volume elements were used for the computational fluid-dynamic analyses. The objective was to investigate the sensitivity of the model to the degree of detail of the µCT image and the elements of the mesh. These findings highlight the pros and cons of the modeling strategy adopted and the importance of such parameters in analyzing fluid behavior in 3D scaffolds.
Collapse
|
27
|
Sánchez-Porras D, Caro-Magdaleno M, González-Gallardo C, García-García ÓD, Garzón I, Carriel V, Campos F, Alaminos M. Generation of a Biomimetic Substitute of the Corneal Limbus Using Decellularized Scaffolds. Pharmaceutics 2021; 13:1718. [PMID: 34684011 PMCID: PMC8541096 DOI: 10.3390/pharmaceutics13101718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022] Open
Abstract
Patients with severe limbal damage and limbal stem cell deficiency are a therapeutic challenge. We evaluated four decellularization protocols applied to the full-thickness and half-thickness porcine limbus, and we used two cell types to recellularize the decellularized limbi. The results demonstrated that all protocols achieved efficient decellularization. However, the method that best preserved the transparency and composition of the limbus extracellular matrix was the use of 0.1% SDS applied to the half-thickness limbus. Recellularization with the limbal epithelial cell line SIRC and human adipose-derived mesenchymal stem cells (hADSCs) was able to generate a stratified epithelium able to express the limbal markers p63, pancytokeratin, and crystallin Z from day 7 in the case of SIRC and after 14-21 days of induction when hADSCs were used. Laminin and collagen IV expression was detected at the basal lamina of both cell types at days 14 and 21 of follow-up. Compared with control native limbi, tissues recellularized with SIRC showed adequate picrosirius red and alcian blue staining intensity, whereas limbi containing hADSCs showed normal collagen staining intensity. These preliminary results suggested that the limbal substitutes generated in this work share important similarities with the native limbus and could be potentially useful in the future.
Collapse
Affiliation(s)
- David Sánchez-Porras
- Department of Histology and Tissue Engineering Group, Faculty of Medicine, Universidad de Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain; (D.S.-P.); (Ó.D.G.-G.); (I.G.); (V.C.)
| | - Manuel Caro-Magdaleno
- Division of Ophthalmology, University Hospital Virgen Macarena, Universidad de Sevilla, E41009 Seville, Spain;
| | | | - Óscar Darío García-García
- Department of Histology and Tissue Engineering Group, Faculty of Medicine, Universidad de Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain; (D.S.-P.); (Ó.D.G.-G.); (I.G.); (V.C.)
- Doctoral Programme in Biomedicine, Escuela Internacional de Posgrado, Universidad de Granada, E18071 Granada, Spain
| | - Ingrid Garzón
- Department of Histology and Tissue Engineering Group, Faculty of Medicine, Universidad de Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain; (D.S.-P.); (Ó.D.G.-G.); (I.G.); (V.C.)
| | - Víctor Carriel
- Department of Histology and Tissue Engineering Group, Faculty of Medicine, Universidad de Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain; (D.S.-P.); (Ó.D.G.-G.); (I.G.); (V.C.)
| | - Fernando Campos
- Department of Histology and Tissue Engineering Group, Faculty of Medicine, Universidad de Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain; (D.S.-P.); (Ó.D.G.-G.); (I.G.); (V.C.)
| | - Miguel Alaminos
- Department of Histology and Tissue Engineering Group, Faculty of Medicine, Universidad de Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain; (D.S.-P.); (Ó.D.G.-G.); (I.G.); (V.C.)
| |
Collapse
|
28
|
Zhao F, Xiong Y, Ito K, van Rietbergen B, Hofmann S. Porous Geometry Guided Micro-mechanical Environment Within Scaffolds for Cell Mechanobiology Study in Bone Tissue Engineering. Front Bioeng Biotechnol 2021; 9:736489. [PMID: 34595161 PMCID: PMC8476750 DOI: 10.3389/fbioe.2021.736489] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Mechanobiology research is for understanding the role of mechanics in cell physiology and pathology. It will have implications for studying bone physiology and pathology and to guide the strategy for regenerating both the structural and functional features of bone. Mechanobiological studies in vitro apply a dynamic micro-mechanical environment to cells via bioreactors. Porous scaffolds are commonly used for housing the cells in a three-dimensional (3D) culturing environment. Such scaffolds usually have different pore geometries (e.g. with different pore shapes, pore dimensions and porosities). These pore geometries can affect the internal micro-mechanical environment that the cells experience when loaded in the bioreactor. Therefore, to adjust the applied micro-mechanical environment on cells, researchers can tune either the applied load and/or the design of the scaffold pore geometries. This review will provide information on how the micro-mechanical environment (e.g. fluid-induced wall shear stress and mechanical strain) is affected by various scaffold pore geometries within different bioreactors. It shall allow researchers to estimate/quantify the micro-mechanical environment according to the already known pore geometry information, or to find a suitable pore geometry according to the desirable micro-mechanical environment to be applied. Finally, as future work, artificial intelligent - assisted techniques, which can achieve an automatic design of solid porous scaffold geometry for tuning/optimising the micro-mechanical environment are suggested.
Collapse
Affiliation(s)
- Feihu Zhao
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
- Zienkiewicz Centre for Computational Engineering, Faculty of Science and Engineering, Swansea University, Swansea, United Kingdom
| | - Yi Xiong
- School of System Design and Intelligent Manufacturing, Southern University of Science and Technology, Shenzhen, China
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Bert van Rietbergen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
29
|
3D Printing and Bioprinting to Model Bone Cancer: The Role of Materials and Nanoscale Cues in Directing Cell Behavior. Cancers (Basel) 2021; 13:cancers13164065. [PMID: 34439218 PMCID: PMC8391202 DOI: 10.3390/cancers13164065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
Bone cancer, both primary and metastatic, is characterized by a low survival rate. Currently, available models lack in mimicking the complexity of bone, of cancer, and of their microenvironment, leading to poor predictivity. Three-dimensional technologies can help address this need, by developing predictive models that can recapitulate the conditions for cancer development and progression. Among the existing tools to obtain suitable 3D models of bone cancer, 3D printing and bioprinting appear very promising, as they enable combining cells, biomolecules, and biomaterials into organized and complex structures that can reproduce the main characteristic of bone. The challenge is to recapitulate a bone-like microenvironment for analysis of stromal-cancer cell interactions and biological mechanics leading to tumor progression. In this review, existing approaches to obtain in vitro 3D-printed and -bioprinted bone models are discussed, with a focus on the role of biomaterials selection in determining the behavior of the models and its degree of customization. To obtain a reliable 3D bone model, the evaluation of different polymeric matrices and the inclusion of ceramic fillers is of paramount importance, as they help reproduce the behavior of both normal and cancer cells in the bone microenvironment. Open challenges and future perspectives are discussed to solve existing shortcomings and to pave the way for potential development strategies.
Collapse
|
30
|
Shi F, Duan K, Yang Z, Liu Y, Weng J. Improved cell seeding efficiency and cell distribution in porous hydroxyapatite scaffolds by semi-dynamic method. Cell Tissue Bank 2021; 23:313-324. [PMID: 34251541 DOI: 10.1007/s10561-021-09945-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/28/2021] [Indexed: 11/26/2022]
Abstract
Tissue engineering is a promising technique for the repair of bone defects. An efficient and homogeneous distribution of cell seeding into scaffold is a crucial but challenging step in the technique. Murine bone marrow mesenchymal stem cells were seeded into porous hydroxyapatite scaffolds of two morphologies by three methods: static seeding, semi-dynamic seeding, or dynamic perfusion seeding. Seeding efficiency, survival, distribution, and proliferation were quantitatively evaluated. To investigate the performance of the three seeding methods for larger/thicker scaffolds as well as batch seeding of numerous scaffolds, three scaffolds were stacked to form assemblies, and seeding efficiencies and cell distribution were analyzed. The semi-dynamic seeding and static seeding methods produced significantly higher seeding efficiencies, vitalities, and proliferation than did the dynamic perfusion seeding. On the other hand, the semi-dynamic seeding and dynamic perfusion seeding methods resulted in more homogeneous cell distribution than did the static seeding. For stacked scaffold assemblies, the semi-dynamic seeding method also created superior seeding efficiency and longitudinal cell distribution homogeneity. The semi-dynamic seeding method combines the high seeding efficiency of static seeding and satisfactory distribution homogeneity of dynamic seeding while circumventing their disadvantages. It may contribute to improved outcomes of bone tissue engineering.
Collapse
Affiliation(s)
- Feng Shi
- Collaboration and Innovation Center of Tissue Repair Material Engineering Technology, China West Normal University, Nanchong, 637009, Sichuan, China
- College of Life Science, China West Normal University, Nanchong, 637009, Sichuan, China
| | - Ke Duan
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zaijun Yang
- Collaboration and Innovation Center of Tissue Repair Material Engineering Technology, China West Normal University, Nanchong, 637009, Sichuan, China
- College of Life Science, China West Normal University, Nanchong, 637009, Sichuan, China
| | - Yumei Liu
- Collaboration and Innovation Center of Tissue Repair Material Engineering Technology, China West Normal University, Nanchong, 637009, Sichuan, China.
- College of Environmental Science and Engineering, China West Normal University, Nanchong, 637009, Sichuan, China.
| | - Jie Weng
- China Key Laboratory of Advanced Technologies of Materials, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| |
Collapse
|
31
|
Moradkhani M, Vahidi B, Ahmadian B. Finite element study of stem cells under fluid flow for mechanoregulation toward osteochondral cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:84. [PMID: 34236534 PMCID: PMC8266696 DOI: 10.1007/s10856-021-06545-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/31/2021] [Indexed: 06/12/2023]
Abstract
Investigating the effects of mechanical stimuli on stem cells under in vitro and in vivo conditions is a very important issue to reach better control on cellular responses like growth, proliferation, and differentiation. In this regard, studying the effects of scaffold geometry, steady, and transient fluid flow, as well as influence of different locations of the cells lodged on the scaffold on effective mechanical stimulations of the stem cells are of the main goals of this study. For this purpose, collagen-based scaffolds and implicit surfaces of the pore architecture was used. In this study, computational fluid dynamics and fluid-structure interaction method was used for the computational simulation. The results showed that the scaffold microstructure and the pore architecture had an essential effect on accessibility of the fluid to different portions of the scaffold. This leads to the optimization of shear stress and hydrodynamic pressure in different surfaces of the scaffold for better transportation of oxygen and growth factors as well as for optimized mechanoregulative responses of cell-scaffold interactions. Furthermore, the results indicated that the HP scaffold provides more optimizer surfaces to culture stem cells rather than Gyroid and IWP scaffolds. The results of exerting oscillatory fluid flow into the HP scaffold showed that the whole surface of the HP scaffold expose to the shear stress between 0.1 and 40 mPa and hydrodynamics factors on the scaffold was uniform. The results of this study could be used as an aid for experimentalists to choose optimist fluid flow conditions and suitable situation for cell culture.
Collapse
Affiliation(s)
- Mehdi Moradkhani
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Bahman Vahidi
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran.
| | - Bahram Ahmadian
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| |
Collapse
|
32
|
Zhang B, Huang J, Narayan RJ. Gradient scaffolds for osteochondral tissue engineering and regeneration. J Mater Chem B 2021; 8:8149-8170. [PMID: 32776030 DOI: 10.1039/d0tb00688b] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The tissue engineering approach for repairing osteochondral (OC) defects involves the fabrication of a biological tissue scaffold that mimics the physiological properties of natural OC tissue (e.g., the gradient transition between the cartilage surface and the subchondral bone). The OC tissue scaffolds described in many research studies exhibit a discrete gradient (e.g., a biphasic or tri/multiphasic structure) or a continuous gradient to mimic OC tissue attributes such as biochemical composition, structure, and mechanical properties. One advantage of a continuous gradient scaffold over biphasic or tri/multiphasic tissue scaffolds is that it more closely mimics natural OC tissue since there is no distinct interface between each layer. Although research studies to this point have yielded good results related to OC regeneration with tissue scaffolds, differences between engineered scaffolds and natural OC tissue remain; due to these differences, current clinical therapies to repair OC defects with engineered scaffolds have not been successful. This paper provides an overview of both discrete and continuous gradient OC tissue scaffolds in terms of cell type, scaffold material, microscale structure, mechanical properties, fabrication methods, and scaffold stimuli. Fabrication of gradient scaffolds with three-dimensional (3D) printing is given special emphasis due to its ability to accurately control scaffold pore geometry. Moreover, the application of computational modeling in OC tissue engineering is considered; for example, efforts to optimize the scaffold structure, mechanical properties, and physical stimuli generated within the scaffold-bioreactor system to predict tissue regeneration are considered. Finally, challenges associated with the repair of OC defects and recommendations for future directions in OC tissue regeneration are proposed.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Mechanical Engineering, University College London, London, UK.
| | - Jie Huang
- Department of Mechanical Engineering, University College London, London, UK.
| | - Roger J Narayan
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
33
|
Ruiz-Alonso S, Lafuente-Merchan M, Ciriza J, Saenz-Del-Burgo L, Pedraz JL. Tendon tissue engineering: Cells, growth factors, scaffolds and production techniques. J Control Release 2021; 333:448-486. [PMID: 33811983 DOI: 10.1016/j.jconrel.2021.03.040] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
Tendon injuries are a global health problem that affects millions of people annually. The properties of tendons make their natural rehabilitation a very complex and long-lasting process. Thanks to the development of the fields of biomaterials, bioengineering and cell biology, a new discipline has emerged, tissue engineering. Within this discipline, diverse approaches have been proposed. The obtained results turn out to be promising, as increasingly more complex and natural tendon-like structures are obtained. In this review, the nature of the tendon and the conventional treatments that have been applied so far are underlined. Then, a comparison between the different tendon tissue engineering approaches that have been proposed to date is made, focusing on each of the elements necessary to obtain the structures that allow adequate regeneration of the tendon: growth factors, cells, scaffolds and techniques for scaffold development. The analysis of all these aspects allows understanding, in a global way, the effect that each element used in the regeneration of the tendon has and, thus, clarify the possible future approaches by making new combinations of materials, designs, cells and bioactive molecules to achieve a personalized regeneration of a functional tendon.
Collapse
Affiliation(s)
- Sandra Ruiz-Alonso
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain
| | - Markel Lafuente-Merchan
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain
| | - Jesús Ciriza
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Laura Saenz-Del-Burgo
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain.
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain.
| |
Collapse
|
34
|
Zhao Q, Zhou Y, Wang M. Three-dimensional endothelial cell incorporation within bioactive nanofibrous scaffolds through concurrent emulsion electrospinning and coaxial cell electrospraying. Acta Biomater 2021; 123:312-324. [PMID: 33508508 DOI: 10.1016/j.actbio.2021.01.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022]
Abstract
Nanofibrous scaffolds hold great promise in tissue engineering owing to their extracellular matrix (ECM)-mimicking architectures. Electrospinning, with its ease for producing nanofibrous scaffolds, has therefore been widely employed for various tissue engineering applications. However, electrospun nanofibrous scaffolds have faced the inherent challenge of three-dimensional (3D) cell distribution due to the small sizes of interconnected pores in these scaffolds when conventional approach of scaffold fabrication with subsequent cell seeding is adopted, which severely limits their applications in repairing/regenerating human body tissues with thick and vascularized structures. In this study, we demonstrate a method to directly place living endothelial cells within bioactive nanofibrous scaffolds in 3D through concurrent emulsion electrospinning and coaxial cell electrospraying. Using this concurrent manufacturing method, endothelial cells are encapsulated in hydrogel microspheres and deposited along with vascular endothelial growth factor (VEGF)-containing nanofibers in the scaffold fabrication process, resulting in nanofibrous scaffolds with 3D embedded cell-encapsulated microspheres. After selective disruption of the hydrogel microspheres, the encapsulated endothelial cells are released, yielding bioactive nanofibrous scaffolds with tissue-like 3D cell-incorporated nanofibrous structures. It is shown that cell viability is well preserved (>98%) during the concurrent manufacturing process and that a deep cell distribution (~100 μm) through the scaffold thickness has been achieved. With combined structural and biochemical cues via the 3D cell-incorporated architectures, endothelial cells can freely stretch, display enhanced intercellular connections, and maintain the phenotype in the bioactive nanofibrous scaffolds. Our investigations offer a promising platform technology for creating bioactive nanofibrous scaffolds with 3D cell incorporation and for overcoming inherent problems of electrospun nanofibrous scaffolds, which should open new avenues for biomanufacturing tissue-mimicking constructs with vascularized structures and complex anatomy. STATEMENT OF SIGNIFICANCE: Electrospun nanofibrous scaffolds face challenges in three-dimensional (3D) cell incorporation and vascularization. Enhancing cell penetration via enlarged interconnected pores is a common strategy to address that. However, there are conflicts between cell penetration and structural integrity for scaffolds formed using such strategy, as deep cell penetration, if possible, can only achieve in highly loose architectures. In this investigation, we demonstrate a concurrent emulsion electrospinning and coaxial cell electrospraying technique, realizing 3D endothelial cell incorporation in electrospun nanofibrous scaffolds independent of cell penetration. Our technology appropriately addresses the conflict between deep 3D cell incorporation and structural integrity. In the scaffolds, the 3D incorporated endothelial cells show well-preserved viability, phenotype and functions, implying improved vascularization potential.
Collapse
|
35
|
Simeoni RB, Mogharbel BF, Francisco JC, Miyague NI, Irioda AC, Souza CMCO, Souza D, Stricker PEF, da Rosa NN, Souza CF, Franco CRC, Sierakowski MR, Abdelwaid E, Guarita-Souza LC, Carvalho KA. Beneficial Roles of Cellulose Patch-Mediated Cell Therapy in Myocardial Infarction: A Preclinical Study. Cells 2021; 10:424. [PMID: 33671407 PMCID: PMC7922134 DOI: 10.3390/cells10020424] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/04/2021] [Accepted: 02/10/2021] [Indexed: 12/27/2022] Open
Abstract
Biological scaffolds have become an attractive approach for repairing the infarcted myocardium and have been shown to facilitate constructive remodeling in injured tissues. This study aimed to investigate the possible utilization of bacterial cellulose (BC) membrane patches containing cocultured cells to limit myocardial postinfarction pathology. Myocardial infarction (MI) was induced by ligating the left anterior descending coronary artery in 45 Wistar rats, and patches with or without cells were attached to the hearts. After one week, the animals underwent echocardiography to assess for ejection fraction and left ventricular end-diastolic and end-systolic volumes. Following patch formation, the cocultured cells retained viability of >90% over 14 days in culture. The patch was applied to the myocardial surface of the infarcted area after staying 14 days in culture. Interestingly, the BC membrane without cellular treatment showed higher preservation of cardiac dimensions; however, we did not observe improvement in the left ventricular ejection fraction of this group compared to coculture-treated membranes. Our results demonstrated an important role for BC in supporting cells known to produce cardioprotective soluble factors and may thus provide effective future therapeutic outcomes for patients suffering from ischemic heart disease.
Collapse
Affiliation(s)
- Rossana B. Simeoni
- Experimental Laboratory of Institute of Biological and Health Sciences of Pontifical Catholic University of Paraná (PUCPR), Street Imaculada Conceição, 1155, 80215-901 Curitiba, Paraná, Brazil; (R.B.S.); (J.C.F.); (N.I.M.); (L.C.G.-S.)
| | - Bassam F. Mogharbel
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Ave., Silva Jardim, 1632, 80240-020 Curitiba, Paraná, Brazil; (B.F.M.); (A.C.I.); (C.M.C.O.S.); (D.S.); (P.E.F.S.); (N.N.d.R.)
| | - Julio C. Francisco
- Experimental Laboratory of Institute of Biological and Health Sciences of Pontifical Catholic University of Paraná (PUCPR), Street Imaculada Conceição, 1155, 80215-901 Curitiba, Paraná, Brazil; (R.B.S.); (J.C.F.); (N.I.M.); (L.C.G.-S.)
| | - Nelson I. Miyague
- Experimental Laboratory of Institute of Biological and Health Sciences of Pontifical Catholic University of Paraná (PUCPR), Street Imaculada Conceição, 1155, 80215-901 Curitiba, Paraná, Brazil; (R.B.S.); (J.C.F.); (N.I.M.); (L.C.G.-S.)
| | - Ana C. Irioda
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Ave., Silva Jardim, 1632, 80240-020 Curitiba, Paraná, Brazil; (B.F.M.); (A.C.I.); (C.M.C.O.S.); (D.S.); (P.E.F.S.); (N.N.d.R.)
| | - Carolina M. C. O. Souza
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Ave., Silva Jardim, 1632, 80240-020 Curitiba, Paraná, Brazil; (B.F.M.); (A.C.I.); (C.M.C.O.S.); (D.S.); (P.E.F.S.); (N.N.d.R.)
| | - Daiany Souza
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Ave., Silva Jardim, 1632, 80240-020 Curitiba, Paraná, Brazil; (B.F.M.); (A.C.I.); (C.M.C.O.S.); (D.S.); (P.E.F.S.); (N.N.d.R.)
| | - Priscila E. Ferreira Stricker
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Ave., Silva Jardim, 1632, 80240-020 Curitiba, Paraná, Brazil; (B.F.M.); (A.C.I.); (C.M.C.O.S.); (D.S.); (P.E.F.S.); (N.N.d.R.)
| | - Nádia Nascimento da Rosa
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Ave., Silva Jardim, 1632, 80240-020 Curitiba, Paraná, Brazil; (B.F.M.); (A.C.I.); (C.M.C.O.S.); (D.S.); (P.E.F.S.); (N.N.d.R.)
| | - Clayton F. Souza
- Biopol, Chemistry Department, Federal University of Paraná, Avenue Cel. Francisco Heráclito dos Santos, 200, 81530-900 Curitiba, Paraná, Brazil; (C.F.S.); (M.-R.S.)
- Chemistry Undergraduate Program, School of Education and Humanities of Pontifical Catholic University of Paraná (PUCPR), Street Imaculada Conceição, 1155, 80215-901 Curitiba, Paraná, Brazil
| | - Celia R. Cavichiolo Franco
- Molecular Biology Department, Federal University of Paraná, Avenue Cel. Francisco Heráclito dos Santos, 100, 81530-900 Curitiba, Paraná, Brazil;
| | - Maria-Rita Sierakowski
- Biopol, Chemistry Department, Federal University of Paraná, Avenue Cel. Francisco Heráclito dos Santos, 200, 81530-900 Curitiba, Paraná, Brazil; (C.F.S.); (M.-R.S.)
| | - Eltyeb Abdelwaid
- Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University, 303 E. Chicago Ave., Tarry 14–725, Chicago, IL 60611, USA;
| | - Luiz C. Guarita-Souza
- Experimental Laboratory of Institute of Biological and Health Sciences of Pontifical Catholic University of Paraná (PUCPR), Street Imaculada Conceição, 1155, 80215-901 Curitiba, Paraná, Brazil; (R.B.S.); (J.C.F.); (N.I.M.); (L.C.G.-S.)
| | - Katherine A.T. Carvalho
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Ave., Silva Jardim, 1632, 80240-020 Curitiba, Paraná, Brazil; (B.F.M.); (A.C.I.); (C.M.C.O.S.); (D.S.); (P.E.F.S.); (N.N.d.R.)
| |
Collapse
|
36
|
Mainardi VL, Arrigoni C, Bianchi E, Talò G, Delcogliano M, Candrian C, Dubini G, Levi M, Moretti M. Improving cell seeding efficiency through modification of fiber geometry in 3D printed scaffolds. Biofabrication 2021; 13. [PMID: 33578401 DOI: 10.1088/1758-5090/abe5b4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/12/2021] [Indexed: 11/11/2022]
Abstract
Cell seeding on 3D scaffolds is a very delicate step in tissue engineering applications, influencing the outcome of the subsequent culture phase, and determining the results of the entire experiment. Thus, it is crucial to maximize its efficiency. To this purpose, a detailed study of the influence of the geometry of the scaffold fibers on dynamic seeding efficiency is presented. 3D printing technology was used to realize PLA porous scaffolds, formed by fibers with a non-circular cross-sectional geometry, named multilobed to highlight the presence of niches and ridges. An oscillating perfusion bioreactor was used to perform bidirectional dynamic seeding of MG63 cells. The fiber shape influences the fluid dynamic parameters of the flow, affecting values of fluid velocity and wall shear stress. The path followed by cells through the scaffold fibers is also affected and results in a larger number of adhered cells in multilobed scaffolds compared to scaffolds with standard pseudo cylindrical fibers. Geometrical and fluid dynamic features can also have an influence on the morphology of adhered cells. The obtained results suggest that the reciprocal influence of geometrical and fluid dynamic features and their combined effect on cell trajectories should be considered to improve the dynamic seeding efficiency when designing scaffold architecture.
Collapse
Affiliation(s)
- Valerio Luca Mainardi
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| | - Chiara Arrigoni
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| | - Elena Bianchi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, ITALY
| | - Giuseppe Talò
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Via Riccardo Galeazzi, 4, Milano, 20161, ITALY
| | - Marco Delcogliano
- Unità di Traumatologia e Ortopedia, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| | - Christian Candrian
- Unità di Traumatologia e Ortopedia, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| | - Gabriele Dubini
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, ITALY
| | - Marinella Levi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, ITALY
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| |
Collapse
|
37
|
Deng F, Liu L, Li Z, Liu J. 3D printed Ti6Al4V bone scaffolds with different pore structure effects on bone ingrowth. J Biol Eng 2021; 15:4. [PMID: 33478505 PMCID: PMC7818551 DOI: 10.1186/s13036-021-00255-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/03/2021] [Indexed: 12/14/2022] Open
Abstract
The microstructure of porous scaffolds plays a vital role in bone regeneration, but its optimal shape is still unclear. In this study, four kinds of porous titanium alloy scaffolds with similar porosities (65%) and pore sizes (650 μm) and different structures were prepared by selective laser melting. Four scaffolds were implanted into the distal femur of rabbits to evaluate bone tissue growth in vivo. Micro-CT and hard tissue section analyses were performed 6 and 12 weeks after the operation to reveal the bone growth of the porous scaffold. The results show that diamond lattice unit (DIA) bone growth is the best of the four topological scaffolds. Through computational fluid dynamics (CFD) analysis, the permeability, velocity and flow trajectory inside the scaffold structure were calculated. The internal fluid velocity difference of the DIA structure is the smallest, and the trajectory of fluid flow inside the scaffold is the longest, which is beneficial for blood vessel growth, nutrient transport and bone formation. In this study, the mechanism of bone growth in different structures was revealed by in vivo experiments combined with CFD, providing a new theoretical basis for the design of bone scaffolds in the future.
Collapse
Affiliation(s)
- Fuyuan Deng
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.,Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, 646000, Sichuan, China
| | - Linlin Liu
- School of Mechanical Engineering, Sichuan University, Chengdu, 610065, Sichuan, China
| | - Zhong Li
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China. .,Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, 646000, Sichuan, China.
| | - Juncai Liu
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China. .,Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
38
|
Pires T, Santos J, Ruben RB, Gouveia BP, Castro APG, Fernandes PR. Numerical-experimental analysis of the permeability-porosity relationship in triply periodic minimal surfaces scaffolds. J Biomech 2021; 117:110263. [PMID: 33493715 DOI: 10.1016/j.jbiomech.2021.110263] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 01/19/2023]
Abstract
Bone Tissue Engineering has been focusing on improving the current methods for bone repair, being the use of scaffolds presented as an upgrade to traditional surgery techniques. Scaffolds are artificially porous matrices, meant to promote cell seeding and proliferation, being these properties influenced by the permeability of the structure. This work employed experimental pressure drop tests and Computational Fluid Dynamics models to assess permeability (and fluid streamlines) within different triply periodic minimal surfaces scaffold geometries (Schwarz D, Gyroid and Schwarz P). The pressure outputs from the computational analysis presented a good correlation with the experimental results, with R2 equal to 0.903; they have also shown that a lower porosity may not mean a lower permeability if the geometry is altered, such as the difference between 60% porous Gyroid scaffolds (8.1*10-9 mm2) and 70% porous Schwarz D scaffolds (7.1*10-9 mm2). Fluid streamlines revealed how the Gyroid geometries are the most appropriate design for most bone tissue engineering applications, due to their consistent fluid permeation, followed by Schwarz D. The Schwarz P geometries have shown flat streamlines and significant variation of the permeability with the porosity (an increase of 10% in their porosity lead to an increase in the permeability from 5.1*10-9 mm2 to 11.7*10-9 mm2), which would imply a poor environment for cell seeding and proliferation.
Collapse
Affiliation(s)
- Tiago Pires
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Jorge Santos
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Rui B Ruben
- ESTG, CDRSP, Polytechnic Institute of Leiria, Leiria, Portugal
| | - Bárbara P Gouveia
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - André P G Castro
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - Paulo R Fernandes
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
39
|
Modulatory properties of extracellular matrix glycosaminoglycans and proteoglycans on neural stem cells behavior: Highlights on regenerative potential and bioactivity. Int J Biol Macromol 2021; 171:366-381. [PMID: 33422514 DOI: 10.1016/j.ijbiomac.2021.01.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/01/2021] [Accepted: 01/02/2021] [Indexed: 12/25/2022]
Abstract
Despite the poor regenerative capacity of the adult central nervous system (CNS) in mammals, two distinct regions, subventricular zone (SVZ) and the subgranular zone (SGZ), continue to generate new functional neurons throughout life which integrate into the pre-existing neuronal circuitry. This process is not fixed but highly modulated, revealing many intrinsic and extrinsic mechanisms by which this performance can be optimized for a given environment. The capacity for self-renewal, proliferation, migration, and multi-lineage potency of neural stem cells (NSCs) underlines the necessity of controlling stem cell fate. In this context, the native and local microenvironment plays a critical role, and the application of this highly organized architecture in the CNS has been considered as a fundamental concept in the generation of new effective therapeutic strategies in tissue engineering approaches. The brain extracellular matrix (ECM) is composed of biomacromolecules, including glycosaminoglycans, proteoglycans, and glycoproteins that provide various biological actions through biophysical and biochemical signaling pathways. Herein, we review predominantly the structure and function of the mentioned ECM composition and their regulatory impact on multiple and diversity of biological functions, including neural regeneration, survival, migration, differentiation, and final destiny of NSCs.
Collapse
|
40
|
Prochor P, Gryko A. Numerical Analysis of the Influence of Porosity and Pore Geometry on Functionality of Scaffolds Designated for Orthopedic Regenerative Medicine. MATERIALS 2020; 14:ma14010109. [PMID: 33383866 PMCID: PMC7796183 DOI: 10.3390/ma14010109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Scaffolds are vital for orthopedic regenerative medicine. Therefore, comprehensive studies evaluating their functionality with consideration of variable parameters are needed. The research aim was to evaluate pore geometry and scaffold porosity influence on first, cell culture efficiency in a perfusion bioreactor and second, osteogenic cell diffusion after its implantation. METHODS For the studies, five pore geometries were selected (triangular prism with a rounded and a flat profile, cube, octagonal prism, sphere) and seven porosities (up to 80%), on the basis of which 70 models were created for finite element analyses. First, scaffolds were placed inside a flow channel to estimate growth medium velocity and wall shear stress. Secondly, scaffolds were placed in a bone to evaluate osteogenic cell diffusion. RESULTS In terms of fluid minimal velocity (0.005 m/s) and maximal wall shear stress (100 mPa), only cubic and octagonal pores with 30% porosity and spherical pores with 20% porosity fulfilled the requirements. Spherical pores had the highest osteogenic cell diffusion efficiency for porosities up to 30%. For higher porosities, the octagonal prism's pores gave the best results up to 80%, where no differences were noted. CONCLUSIONS The data obtained allows for the appropriate selection of pore geometry and scaffold porosity for orthopedic regenerative medicine.
Collapse
|
41
|
Di Bartolo A, Melchels FPW. Prolonged recovery of 3D printed, photo-cured polylactide shape memory polymer networks. APL Bioeng 2020; 4:036105. [PMID: 32844139 PMCID: PMC7442493 DOI: 10.1063/5.0008910] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 08/07/2020] [Indexed: 01/29/2023] Open
Abstract
Shape memory polymers are materials that are able to retain a deformed state until an external stimulus, most typically heat, triggers recovery to the original geometry. Whereas typically, shape memory polymers are required to recover fast (seconds to minutes), many applications, particularly in the medical field, would benefit from a slow recovery (days to weeks). In this work, we exploit the broad glass transition range of photo-cured poly(D,L-lactide) dimethacrylate networks to obtain recovery times of up to 2 weeks, at 11 °C below the peak glass transition temperature of 58 °C. Recovery times decreased considerably for higher recovery temperatures, down to ∼10 min at 55 °C. A large spread in glass transition values (53.3-61.0 °C) was observed between samples, indicating poor reproducibility in sample preparation and, hence, in predicting shape recovery kinetics for individual samples. Furthermore, a staged recovery was observed with different parts of the samples recovering at different times. The ability to prepare complex structures using digital light processing stereolithography 3D printing from these polymers was confirmed. To the best of our knowledge, this work provides the first experimental evidence of prolonged recovery of shape memory polymers.
Collapse
Affiliation(s)
- Alberto Di Bartolo
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, EH14 4AS Edinburgh, United Kingdom
| | - Ferry P. W. Melchels
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, EH14 4AS Edinburgh, United Kingdom
| |
Collapse
|
42
|
Puppi D, Chiellini F. Biodegradable Polymers for Biomedical Additive Manufacturing. APPLIED MATERIALS TODAY 2020; 20:100700. [DOI: 10.1016/j.apmt.2020.100700] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
43
|
Charbonnier B, Manassero M, Bourguignon M, Decambron A, El-Hafci H, Morin C, Leon D, Bensidoum M, Corsia S, Petite H, Marchat D, Potier E. Custom-made macroporous bioceramic implants based on triply-periodic minimal surfaces for bone defects in load-bearing sites. Acta Biomater 2020; 109:254-266. [PMID: 32194263 DOI: 10.1016/j.actbio.2020.03.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022]
Abstract
The architectural features of synthetic bone grafts are key parameters for regulating cell functions and tissue formation for the successful repair of bone defects. In this regard, macroporous structures based on triply-periodic minimal surfaces (TPMS) are considered to have untapped potential. In the present study, custom-made implants based on a gyroid structure, with (GPRC) and without (GP) a cortical-like reinforcement, were specifically designed to fit an intended bone defect in rat femurs. Sintered hydroxyapatite implants were produced using a dedicated additive manufacturing technology and their morphological, physico-chemical and mechanical features were characterized. The implants' integrity and ability to support bone ingrowth were assessed after 4, 6 and 8 weeks of implantation in a 3-mm-long, femoral defect in Lewis rats. GP and GPRC implants were manufactured with comparable macro- to nano-architectures. Cortical-like reinforcement significantly improved implant effective stiffness and resistance to fracture after implantation. This cortical-like reinforcement also concentrated new bone formation in the core of the GPRC implants, without affecting newly formed bone quantity or maturity. This study showed, for the first time, that custom-made TPMS-based bioceramic implants could be produced and successfully implanted in load-bearing sites. Adding a cortical-like reinforcement (GPRC implants) was a relevant solution to improve implant mechanical resistance, and changed osteogenic mechanism compared to the GP implants. STATEMENT OF SIGNIFICANCE: Architectural features are known to be key parameters for successful bone repair using synthetic bioceramic bone graft. So far, conventional manufacturing techniques, lacking reproducibility and complete control of the implant macro-architecture, impeded the exploration of complex architectures, such as triply periodic minimal surfaces (TPMS), which are foreseen to have an unrivaled potential for bone repair. Using a new additive manufacturing process, macroporous TPMS-based bioceramics implants were produced in calcium phosphate, characterized and implanted in a femoral defect in rats. The results showed, for the first time, that such macroporous implants can be successfully implanted in anatomical load-bearing sites when a cortical-like outer shell is added. This outer shell also concentrated new bone formation in the implant center, without affecting new bone quantity or maturity.
Collapse
|
44
|
Conrad B, Hayashi C, Yang F. Gelatin-Based Microribbon Hydrogels Support Robust MSC Osteogenesis across a Broad Range of Stiffness. ACS Biomater Sci Eng 2020; 6:3454-3463. [PMID: 33463171 PMCID: PMC10154176 DOI: 10.1021/acsbiomaterials.9b01792] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Scaffold macroporosity has been shown to be critical for promoting bone regeneration. Although injectable materials are preferred for minimally invasive delivery, conventional macroporous scaffolds were not injectable and do not support homogeneous cell encapsulation. We recently reported a gelatin-based microribbon (μRB) scaffold that offers macroporosity while also supporting homogeneous cell encapsulation. Compared to conventional gelatin hydrogels, macroporous gelatin μRB scaffolds demonstrated great advantage in enhancing mesenchymal stem cell (MSC)-based cartilage formation. However, whether gelatin-based μRBs support MSC osteogenesis and bone formation remains unknown. The goal of this study is to assess the potential of gelatin-based μRBs for supporting MSC-based osteogenesis and bone formation in vitro. Given recent evidence from the literature that osteogenesis is sensitive to substrate stiffness, we further investigate how varying μRB stiffness modulates MSC osteogenesis. We first determine the maximal stiffness range of gelatin μRBs that can be fabricated (13-57 kPa), which supports both retention of μRB shape and macroporosity within scaffolds after inter-cross-linking. Interestingly, varying μRB stiffness across a broad range of stiffness did not significantly impact osteogenesis, with all groups supporting upregulation of bone markers and extensive collagen deposition. All gelatin μRBs also supported a comparable level of cell spreading and upregulation of mechanosensing markers. However, soft μRB (13 kPa) scaffolds did not maintain structural integrity and condensed into a pellet over time. Both intermediate and stiff gelatin μRB-based scaffolds maintained their integrity and supported robust bone formation, leading to a more than 10-fold increase in the compressive moduli of engineered bone after 5 weeks of culture in osteogenic media. Incorporating hydroxyapatite (HA) nanoparticle coating onto the gelatin μRB surface further accelerated the maturation of MSCs into osteoblasts and mineralization. Together, these results validate that gelatin μRBs can support MSC osteogenesis across a broad range of stiffness and offers an injectable macroporous scaffold for enhancing stem-cell-based bone regeneration.
Collapse
Affiliation(s)
- Bogdan Conrad
- Program of Stem Cell Biology and Regenerative Medicine, Stanford University, 300 Pasteur Drive, Edward Building Room 114, Stanford, California94305, United States
| | - Camila Hayashi
- Department of Chemical Engineering, Stanford University Shriram Center, Room 129, Stanford, California94305, United States
| | - Fan Yang
- Department of Orthopaedic Surgery Department of Bioengineering, Stanford University300 Pasteur Drive, Edward Building Room 114, Stanford, California94305, United States
| |
Collapse
|
45
|
Zhang B, Cristescu R, Chrisey DB, Narayan RJ. Solvent-based Extrusion 3D Printing for the Fabrication of Tissue Engineering Scaffolds. Int J Bioprint 2020; 6:211. [PMID: 32596549 PMCID: PMC7294686 DOI: 10.18063/ijb.v6i1.211] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 12/02/2019] [Indexed: 11/23/2022] Open
Abstract
Three-dimensional (3D) printing has been emerging as a new technology for scaffold fabrication to overcome the problems associated with the undesirable microstructure associated with the use of traditional methods. Solvent-based extrusion (SBE) 3D printing is a popular 3D printing method, which enables incorporation of cells during the scaffold printing process. The scaffold can be customized by optimizing the scaffold structure, biomaterial, and cells to mimic the properties of natural tissue. However, several technical challenges prevent SBE 3D printing from translation to clinical use, such as the properties of current biomaterials, the difficulties associated with simultaneous control of multiple biomaterials and cells, and the scaffold-to-scaffold variability of current 3D printed scaffolds. In this review paper, a summary of SBE 3D printing for tissue engineering (TE) is provided. The influences of parameters such as ink biomaterials, ink rheological behavior, cross-linking mechanisms, and printing parameters on scaffold fabrication are considered. The printed scaffold structure, mechanical properties, degradation, and biocompatibility of the scaffolds are summarized. It is believed that a better understanding of the scaffold fabrication process and assessment methods can improve the functionality of SBE-manufactured 3D printed scaffolds.
Collapse
Affiliation(s)
- Bin Zhang
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| | - Rodica Cristescu
- National Institute for Lasers, Plasma and Radiation Physics, Lasers Department, P.O. Box MG-36, Bucharest-Magurele, Romania
| | - Douglas B Chrisey
- Department of Physics and Engineering Physics, Tulane University, New Orleans, LA, USA
| | - Roger J Narayan
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| |
Collapse
|
46
|
A 3D computational model of perfusion seeding for investigating cell transport and adhesion within a porous scaffold. Biomech Model Mechanobiol 2020; 19:1461-1475. [PMID: 31900653 DOI: 10.1007/s10237-019-01281-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/17/2019] [Indexed: 10/25/2022]
Abstract
The process of cell seeding within a porous scaffold is an essential first step in the development of tissue-engineered bone grafts. Understanding the underlying mechanisms of cell distribution and adhesion is fundamental for the design and optimization of the seeding process. To that end, we present a numerical model to investigate the perfusion cell seeding process that incorporates cell mechanics, cell-fluid interaction, and cell-scaffold adhesion. The individual cells are modeled as deformable spherical capsules capable of adhering to the scaffold surface as well as to other cells with probabilistic bond formation and rupture. The mechanical deformation of the cell is calibrated with the stretching of mice mesenchymal stem cells induced by optical tweezers, while the predicted adhesive forces are consistent with the experimental data reported in the literature. A sub-domain is numerically reconstructed as the region of interest (ROI) which is representative of an actual scaffold. Through the simulations, the perfusion seeding kinetics within the ROI involving detailed transport and adhesion of cells over time is analyzed. The effects of the perfusion pressure and initial cell concentration on the seeding kinetics are studied in terms of adhesion rates, cell cluster formation, seeding uniformity, and efficiency, as well as scaffold permeability. The results highlight the importance of cell-fluid interaction and adhesion dynamics in modeling the dynamic seeding process. This bottom-up model provides a way to bridge detailed behaviors of individual cells to the seeding outcomes at the macroscopic scale, allowing for finding the best configuration to enhance cell seeding.
Collapse
|
47
|
Li Y, Jahr H, Pavanram P, Bobbert FSL, Paggi U, Zhang XY, Pouran B, Leeflang MA, Weinans H, Zhou J, Zadpoor AA. Additively manufactured functionally graded biodegradable porous iron. Acta Biomater 2019; 96:646-661. [PMID: 31302295 DOI: 10.1016/j.actbio.2019.07.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 11/28/2022]
Abstract
Additively manufactured (AM) functionally graded porous metallic biomaterials offer unique opportunities to satisfy the contradictory design requirements of an ideal bone substitute. However, no functionally graded porous structures have ever been 3D-printed from biodegradable metals, even though biodegradability is crucial both for full tissue regeneration and for the prevention of implant-associated infections in the long term. Here, we present the first ever report on AM functionally graded biodegradable porous metallic biomaterials. We made use of a diamond unit cell for the topological design of four different types of porous structures including two functionally graded structures and two reference uniform structures. Specimens were then fabricated from pure iron powder using selective laser melting (SLM), followed by experimental and computational analyses of their permeability, dynamic biodegradation behavior, mechanical properties, and cytocompatibility. It was found that the topological design with functional gradients controlled the fluid flow, mass transport properties and biodegradation behavior of the AM porous iron specimens, as up to 4-fold variations in permeability and up to 3-fold variations in biodegradation rate were observed for the different experimental groups. After 4 weeks of in vitro biodegradation, the AM porous scaffolds lost 5-16% of their weight. This falls into the desired range of biodegradation rates for bone substitution and confirms our hypothesis that topological design could indeed accelerate the biodegradation of otherwise slowly degrading metals, like iron. Even after 4 weeks of biodegradation, the mechanical properties of the specimens (i.e., E = 0.5-2.1 GPa, σy = 8-48 MPa) remained within the range of the values reported for trabecular bone. Design-dependent cell viability did not differ from gold standard controls for up to 48 h. This study clearly shows the great potential of AM functionally graded porous iron as a bone substituting material. Moreover, we demonstrate that complex topological design permits the control of mechanical properties, degradation behavior of AM porous metallic biomaterials. STATEMENT OF SIGNIFICANCE: No functionally graded porous structures have ever been 3D-printed from biodegradable metals, even though biodegradability is crucial both for full tissue regeneration and for the prevention of implant-associated infections in the long term. Here, we present the first report on 3D-printed functionally graded biodegradable porous metallic biomaterials. Our results suggest that topological design in general, and functional gradients in particular can be used as an important tool for adjusting the biodegradation behavior of AM porous metallic biomaterials. The biodegradation rate and mass transport properties of AM porous iron can be increased while maintaining the bone-mimicking mechanical properties of these biomaterials. The observations reported here underline the importance of proper topological design in the development of AM porous biodegradable metals.
Collapse
Affiliation(s)
- Y Li
- Department of Biomechanical Engineering, Delft University of Technology, Delft 2628 CD, The Netherlands.
| | - H Jahr
- Department of Anatomy and Cell Biology, University Hospital RWTH Aachen, Aachen 52074, Germany; Department of Orthopedic Surgery, Maastricht UMC+, Maastricht 6202 AZ, The Netherlands
| | - P Pavanram
- Department of Anatomy and Cell Biology, University Hospital RWTH Aachen, Aachen 52074, Germany
| | - F S L Bobbert
- Department of Biomechanical Engineering, Delft University of Technology, Delft 2628 CD, The Netherlands
| | - U Paggi
- 3D Systems - LayerWise NV, Grauwmeer 14, Leuven 3001, Belgium; KU Leuven Department of Mechanical Engineering, Kasteelpark Arenberg 44, Leuven 3001, Belgium
| | - X-Y Zhang
- Department of Mechanical Engineering, Tsinghua University, Beijing 10004, China
| | - B Pouran
- Department of Biomechanical Engineering, Delft University of Technology, Delft 2628 CD, The Netherlands; Department of Orthopedics, UMC Utrecht, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| | - M A Leeflang
- Department of Biomechanical Engineering, Delft University of Technology, Delft 2628 CD, The Netherlands
| | - H Weinans
- Department of Biomechanical Engineering, Delft University of Technology, Delft 2628 CD, The Netherlands; Department of Orthopedics, UMC Utrecht, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| | - J Zhou
- Department of Biomechanical Engineering, Delft University of Technology, Delft 2628 CD, The Netherlands
| | - A A Zadpoor
- Department of Biomechanical Engineering, Delft University of Technology, Delft 2628 CD, The Netherlands
| |
Collapse
|
48
|
Krause AL, Beliaev D, Van Gorder RA, Waters SL. Lattice and continuum modelling of a bioactive porous tissue scaffold. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2019; 36:325-360. [PMID: 30107530 DOI: 10.1093/imammb/dqy012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 01/18/2018] [Accepted: 07/16/2018] [Indexed: 12/29/2022]
Abstract
A contemporary procedure to grow artificial tissue is to seed cells onto a porous biomaterial scaffold and culture it within a perfusion bioreactor to facilitate the transport of nutrients to growing cells. Typical models of cell growth for tissue engineering applications make use of spatially homogeneous or spatially continuous equations to model cell growth, flow of culture medium, nutrient transport and their interactions. The network structure of the physical porous scaffold is often incorporated through parameters in these models, either phenomenologically or through techniques like mathematical homogenization. We derive a model on a square grid lattice to demonstrate the importance of explicitly modelling the network structure of the porous scaffold and compare results from this model with those from a modified continuum model from the literature. We capture two-way coupling between cell growth and fluid flow by allowing cells to block pores, and by allowing the shear stress of the fluid to affect cell growth and death. We explore a range of parameters for both models and demonstrate quantitative and qualitative differences between predictions from each of these approaches, including spatial pattern formation and local oscillations in cell density present only in the lattice model. These differences suggest that for some parameter regimes, corresponding to specific cell types and scaffold geometries, the lattice model gives qualitatively different model predictions than typical continuum models. Our results inform model selection for bioactive porous tissue scaffolds, aiding in the development of successful tissue engineering experiments and eventually clinically successful technologies.
Collapse
Affiliation(s)
- Andrew L Krause
- Mathematical Institute, Andrew Wiles Building, University of Oxford, Radcliffe Observatory Quarter, Woodstock Rd, UK
| | - Dmitry Beliaev
- Mathematical Institute, Andrew Wiles Building, University of Oxford, Radcliffe Observatory Quarter, Woodstock Rd, UK
| | - Robert A Van Gorder
- Mathematical Institute, Andrew Wiles Building, University of Oxford, Radcliffe Observatory Quarter, Woodstock Rd, UK
| | - Sarah L Waters
- Mathematical Institute, Andrew Wiles Building, University of Oxford, Radcliffe Observatory Quarter, Woodstock Rd, UK
| |
Collapse
|
49
|
Pederzoli F, Joice G, Salonia A, Bivalacqua TJ, Sopko NA. Regenerative and engineered options for urethroplasty. Nat Rev Urol 2019; 16:453-464. [PMID: 31171866 DOI: 10.1038/s41585-019-0198-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
Abstract
Surgical correction of urethral strictures by substitution urethroplasty - the use of grafts or flaps to correct the urethral narrowing - remains one of the most challenging procedures in urology and is frequently associated with complications, restenosis and poor quality of life for the affected individual. Tissue engineering using different cell types and tissue scaffolds offers a promising alternative for tissue repair and replacement. The past 30 years of tissue engineering has resulted in the development of several therapies that are now in use in the clinic, especially in treating cutaneous, bone and cartilage defects. Advances in tissue engineering for urethral replacement have resulted in several clinical applications that have shown promise but have not yet become the standard of care.
Collapse
Affiliation(s)
- Filippo Pederzoli
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Gregory Joice
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Trinity J Bivalacqua
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Nikolai A Sopko
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
50
|
Conrad B, Han LH, Yang F. Gelatin-Based Microribbon Hydrogels Accelerate Cartilage Formation by Mesenchymal Stem Cells in Three Dimensions. Tissue Eng Part A 2019; 24:1631-1640. [PMID: 29926770 DOI: 10.1089/ten.tea.2018.0011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hydrogels (HGs) are attractive matrices for cell-based cartilage tissue regeneration given their injectability and ability to fill defects with irregular shapes. However, most HGs developed to date often lack cell scale macroporosity, which restrains the encapsulated cells, leading to delayed new extracellular matrix deposition restricted to pericellular regions. Furthermore, tissue-engineered cartilage using conventional HGs generally suffers from poor mechanical property and fails to restore the load-bearing property of articular cartilage. The goal of this study was to evaluate the potential of macroporous gelatin-based microribbon (μRB) HGs as novel 3D matrices for accelerating chondrogenesis and new cartilage formation by human mesenchymal stem cells (MSCs) in 3D with improved mechanical properties. Unlike conventional HGs, these μRB HGs are inherently macroporous and exhibit cartilage-mimicking shock-absorbing mechanical property. After 21 days of culture, MSC-seeded μRB scaffolds exhibit a 20-fold increase in compressive modulus to 225 kPa, a range that is approaching the level of native cartilage. In contrast, HGs only resulted in a modest increase in compressive modulus of 65 kPa. Compared with conventional HGs, macroporous μRB scaffolds significantly increased the total amount of neocartilage produced by MSCs in 3D, with improved interconnectivity and mechanical strength. Altogether, these results validate gelatin-based μRBs as promising scaffolds for enhancing and accelerating MSC-based cartilage regeneration and may be used to enhance cartilage regeneration using other cell types as well.
Collapse
Affiliation(s)
- Bogdan Conrad
- 1 Program of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Li-Hsin Han
- 2 Department of Orthopedic Surgery, Stanford University School of Medicine , Stanford, California
| | - Fan Yang
- 3 Department of Orthopedic Surgery and Bioengineering, Stanford University , Stanford, California
| |
Collapse
|