1
|
Hashemi-Afzal F, Fallahi H, Bagheri F, Collins MN, Eslaminejad MB, Seitz H. Advancements in hydrogel design for articular cartilage regeneration: A comprehensive review. Bioact Mater 2025; 43:1-31. [PMID: 39318636 PMCID: PMC11418067 DOI: 10.1016/j.bioactmat.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024] Open
Abstract
This review paper explores the cutting-edge advancements in hydrogel design for articular cartilage regeneration (CR). Articular cartilage (AC) defects are a common occurrence worldwide that can lead to joint breakdown at a later stage of the disease, necessitating immediate intervention to prevent progressive degeneration of cartilage. Decades of research into the biomedical applications of hydrogels have revealed their tremendous potential, particularly in soft tissue engineering, including CR. Hydrogels are highly tunable and can be designed to meet the key criteria needed for a template in CR. This paper aims to identify those criteria, including the hydrogel components, mechanical properties, biodegradability, structural design, and integration capability with the adjacent native tissue and delves into the benefits that CR can obtain through appropriate design. Stratified-structural hydrogels that emulate the native cartilage structure, as well as the impact of environmental stimuli on the regeneration outcome, have also been discussed. By examining recent advances and emerging techniques, this paper offers valuable insights into developing effective hydrogel-based therapies for AC repair.
Collapse
Affiliation(s)
- Fariba Hashemi-Afzal
- Biotechnology Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Hooman Fallahi
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, 14115-111, Iran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104 USA
| | - Fatemeh Bagheri
- Biotechnology Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Maurice N. Collins
- School of Engineering, Bernal Institute and Health Research Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 16635-148, Iran
| | - Hermann Seitz
- Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Justus-von-Liebig-Weg 6, 18059 Rostock, Germany
- Department Life, Light & Matter, University of Rostock, Albert-Einstein-Straße 25, 18059 Rostock, Germany
| |
Collapse
|
2
|
van den Nieuwenhof DWA, Moroni L, Chou J, Hinkelbein J. Cellular response in three-dimensional spheroids and tissues exposed to real and simulated microgravity: a narrative review. NPJ Microgravity 2024; 10:102. [PMID: 39505879 PMCID: PMC11541851 DOI: 10.1038/s41526-024-00442-z] [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: 01/24/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024] Open
Abstract
The rising aging population underscores the need for advances in tissue engineering and regenerative medicine. Alterations in cellular response in microgravity might be pivotal in unraveling the intricate cellular mechanisms governing tissue and organ regeneration. Microgravity could improve multicellular spheroid, tissue, and organ formation. This review summarizes microgravity-induced cellular alterations and highlights the potential of tissue engineering in microgravity for future breakthroughs in space travel, transplantation, drug testing, and personalized medicine.
Collapse
Affiliation(s)
| | - Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Complex Tissue Engineering, Maastricht University, Maastricht, The Netherlands
| | - Joshua Chou
- University of Technology Sydney (UTS), Sydney, NSW, Australia
| | - Jochen Hinkelbein
- Department of Anesthesiology, Intensive Care Medicine and Emergency Medicine, Johannes Wesling Klinikum Minden, University Hospital Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
3
|
Nerger BA, Kashyap K, Deveney BT, Lou J, Hanan BF, Liu Q, Khalil A, Lungjangwa T, Cheriyan M, Gupta A, Jaenisch R, Weitz DA, Mahadevan L, Mooney DJ. Tuning porosity of macroporous hydrogels enables rapid rates of stress relaxation and promotes cell expansion and migration. Proc Natl Acad Sci U S A 2024; 121:e2410806121. [PMID: 39467139 PMCID: PMC11551365 DOI: 10.1073/pnas.2410806121] [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: 05/30/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
Extracellular matrix (ECM) viscoelasticity broadly regulates cell behavior. While hydrogels can approximate the viscoelasticity of native ECM, it remains challenging to recapitulate the rapid stress relaxation observed in many tissues without limiting the mechanical stability of the hydrogel. Here, we develop macroporous alginate hydrogels that have an order of magnitude increase in the rate of stress relaxation as compared to bulk hydrogels. The increased rate of stress relaxation occurs across a wide range of polymer molecular weights (MWs), which enables the use of high MW polymer for improved mechanical stability of the hydrogel. The rate of stress relaxation in macroporous hydrogels depends on the volume fraction of pores and the concentration of bovine serum albumin, which is added to the hydrogels to stabilize the macroporous structure during gelation. Relative to cell spheroids encapsulated in bulk hydrogels, spheroids in macroporous hydrogels have a significantly larger area and smaller circularity because of increased cell migration. A computational model provides a framework for the relationship between the macroporous architecture and morphogenesis of encapsulated spheroids that is consistent with experimental observations. Taken together, these findings elucidate the relationship between macroporous hydrogel architecture and stress relaxation and help to inform the design of macroporous hydrogels for materials-based cell therapies.
Collapse
Affiliation(s)
- Bryan A. Nerger
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| | - Kirti Kashyap
- Department of Physics, Indian Institute of Technology Hyderabad, Sangareddy, Telangana502285, India
| | - Brendan T. Deveney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Junzhe Lou
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| | - Blake F. Hanan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| | - Qi Liu
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
| | - Andrew Khalil
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
| | | | | | - Anupam Gupta
- Department of Physics, Indian Institute of Technology Hyderabad, Sangareddy, Telangana502285, India
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02142
| | - David A. Weitz
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Physics, Harvard University, Cambridge, MA02138
| | - L. Mahadevan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Department of Physics, Harvard University, Cambridge, MA02138
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA02138
| | - David J. Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| |
Collapse
|
4
|
Hayashi Y, Fujii T, Kim S, Ozeki T, Badylak SF, D'Amore A, Mutsuga M, Wagner WR. Intervening to Preserve Function in Ischemic Cardiomyopathy with a Porous Hydrogel and Extracellular Matrix Composite in a Rat Myocardial Infarction Model. Adv Healthc Mater 2024:e2402757. [PMID: 39491520 DOI: 10.1002/adhm.202402757] [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: 07/25/2024] [Revised: 10/10/2024] [Indexed: 11/05/2024]
Abstract
Multiple hydrogels are developed for injection therapy after myocardial infarction, with some incorporating substances promoting tissue regeneration and others emphasizing mechanical effects. In this study, porosity and extracellular matrix-derived digest (ECM) are incorporated, into a mechanically optimized, thermoresponsive, degradable hydrogel (poly(N-isopropylacrylamide-co-N-vinylpyrrolidone-co-MAPLA)) and evaluate whether this biomaterial injectate can abrogate adverse remodeling in rat ischemic cardiomyopathy. After myocardial infarction, rats are divided into four groups: NP (non-porous hydrogel) without either ECM or porosity, PM (porous hydrogel) from the same synthetic copolymer with mannitol beads as porogens, and PME with porosity and ECM digest added to the synthetic copolymer. PBS injection alone is a control group. Intramyocardial injections occurred 3 days after myocardial infarction followed by serial echocardiography and histological assessments 8 weeks after infarction. Echocardiographic function and neovascularization improved in the PME group compared to the other hydrogels and PBS injection. The PME group also demonstrated improved LV geometry and macrophage polarization (toward M2) compared to PBS, whereas differences are not observed in the NP or PM groups versus control. These results demonstrate further functional improvement may be achieved in hydrogel injection therapy for ischemic cardiomyopathy by incorporating porosity and ECM digest, representing combined mechanical and biological effects.
Collapse
Affiliation(s)
- Yasunari Hayashi
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 4668550, Japan
| | - Taro Fujii
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 4668550, Japan
| | - Seungil Kim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Agricultural and Biological Engineering, Mississippi State University, MS, 39762, USA
| | - Takahiro Ozeki
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 4668550, Japan
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Antonio D'Amore
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Fondazione RiMED, Palermo, 90133, Italy
| | - Masato Mutsuga
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 4668550, Japan
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
5
|
Orozco-Osorio YA, Gaita-Anturi AV, Ossa-Orozco CP, Arias-Acevedo M, Uribe D, Paucar C, Vasquez AF, Saldarriaga W, Ramirez JG, Lopera A, García C. Utilization of Additive Manufacturing Techniques for the Development of a Novel Scaffolds with Magnetic Properties for Potential Application in Enhanced Bone Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402419. [PMID: 39004887 DOI: 10.1002/smll.202402419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/24/2024] [Indexed: 07/16/2024]
Abstract
This study focuses on designing and evaluating scaffolds with essential properties for bone regeneration, such as biocompatibility, macroporous geometry, mechanical strength, and magnetic responsiveness. The scaffolds are made using 3D printing with acrylic resin and iron oxides synthesized through solution combustion. Utilizing triply periodic minimal surfaces (TPMS) geometry and mask stereolithography (MSLA) printing, the scaffolds achieve precise geometrical features. The mechanical properties are enhanced through resin curing, and magnetite particles from synthesized nanoparticles and alluvial magnetite are added for magnetic properties. The scaffolds show a balance between stiffness, porosity, and magnetic responsiveness, with maximum compression strength between 4.8 and 9.2 MPa and Young's modulus between 58 and 174 MPa. Magnetic properties such as magnetic coercivity, remanence, and saturation are measured, with the best results from scaffolds containing synthetic iron oxides at 1% weight. The viscosity of the mixtures used for printing is between 350 and 380 mPas, and contact angles between 90° and 110° are achieved. Biocompatibility tests indicate the potential for clinical trials, though further research is needed to understand the impact of magnetic properties on cellular interactions and optimize scaffold design for specific applications. This integrated approach offers a promising avenue for the development of advanced materials capable of promoting enhanced bone regeneration.
Collapse
Affiliation(s)
| | | | | | - María Arias-Acevedo
- Instituto Tecnológico Metropolitano, Calle 73 #76A-354, Campus Robledo, Medellín, Antioquia, 50034, Colombia
| | - Diego Uribe
- Instituto Tecnológico Metropolitano, Calle 73 #76A-354, Campus Robledo, Medellín, Antioquia, 50034, Colombia
| | - Carlos Paucar
- Universidad Nacional de Colombia sede Medellín, Carrera 65 # 59A-100, Medellin, Antioquia, 050034, Colombia
| | | | - Wilmer Saldarriaga
- Universidad Nacional de Colombia sede Medellín, Carrera 65 # 59A-100, Medellin, Antioquia, 050034, Colombia
| | | | - Alex Lopera
- Grupo de Nanoestructuras y Física Aplicada (NANOUPAR), Universidad Nacional de Colombia, La Paz, 202017, Colombia
| | - Claudia García
- Universidad Nacional de Colombia sede Medellín, Carrera 65 # 59A-100, Medellin, Antioquia, 050034, Colombia
| |
Collapse
|
6
|
Perry AC, Adesida AB. Tissue Engineering Nasal Cartilage Grafts with Three-Dimensional Printing: A Comprehensive Review. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39311456 DOI: 10.1089/ten.teb.2024.0187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Nasal cartilage serves a crucial structural function for the nose, where rebuilding the cartilaginous framework is an essential aspect of nasal reconstruction. Conventional methods of nasal reconstruction rely on autologous cartilage harvested from patients, which contributes to donor site pain and the potential for site-specific complications. Some patients are not ideal candidates for this procedure due to a lack of adequate substitute cartilage due to age-related calcification, differences in tissue quality, or due to prior surgeries. Tissue engineering, combined with three-dimensional printing technologies, has emerged as a promising method of generating biomimetic tissues to circumvent these issues to restore normal function and aesthetics. We conducted a comprehensive literature review to examine the applications of three-dimensional printing in conjunction with tissue engineering for the generation of nasal cartilage grafts. This review aims to compare various approaches and discuss critical considerations in the design of these grafts.
Collapse
Affiliation(s)
- Alexander C Perry
- Department of Surgery, Division of Plastic Surgery, University of Alberta, Edmonton, Canada
- Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, University of Alberta, Edmonton, Canada
| | - Adetola B Adesida
- Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, University of Alberta, Edmonton, Canada
- Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, Canada
| |
Collapse
|
7
|
Mandatori D, D'Amico E, Romasco T, Gatto ML, Notarangelo MP, Mangano C, Furlani M, Penolazzi L. A 3D in vitro model of biphasic calcium phosphate (BCP) scaffold combined with human osteoblasts, osteoclasts, and endothelial cells as a platform to mimic the oral microenvironment for tissue regeneration. J Dent 2024; 151:105411. [PMID: 39426560 DOI: 10.1016/j.jdent.2024.105411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVES This study aimed to develop an innovative 3D in vitro model based on the biphasic calcium phosphate (BCP) scaffold combined with human osteoblasts (hOBs), osteoclasts (hOCs), and endothelial cells to evaluate its effects on bone and vascular cells behavior. METHODS To this end, an optimized mixture of hydroxyapatite (HA) and β-tricalcium phosphate (TCP) with a weight ratio of 30/70 was employed to develop a BCP scaffold using the computer-aided design (CAD) approach. The BCP scaffold was combined with primary cultures of hOBs, hOCs and human umbilical vein endothelial cells (HUVECs). RESULTS Morphometric analyses using scanning electron microscopy (SEM) and X-ray micro-computed tomography, along with biomechanical testing, revealed that BCP scaffold exhibited a regular 3D structure with large interconnected internal pores (700 µm) and high mechanical strength. In terms of biological behavior, after 14 days of tri-culture with hOBs, hMCs and HUVECs, SEM, immunofluorescence, and histological analyses showed that all cell types were viable and adhered well to the entire surface of the scaffold. Interestingly, SEM and energy-dispersive X-ray spectroscopy analyses also revealed on the BCP scaffold the presence of mineralized matrix crystals of Ca, P, O and C within a tissue-like cell layer produced by the interaction of the three cell types. CONCLUSIONS Data confirmed the high performance of the BCP scaffold through biomechanical studies. Notably, for the first time, this study demonstrated the feasibility of combining BCP scaffold with hOBs, hOCs, and HUVEC, which remained viable and maintained their native phenotypes, creating also tissue-like cell layer. CLINICAL SIGNIFICANCE Although further investigation is needed, these results underscore the potential to develop a 3D in vitro model that mimics the oral microenvironment, which could be valuable for BTE approaches in vivo studies.
Collapse
Affiliation(s)
- Domitilla Mandatori
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology CAST, "G. d' Annunzio" University Chieti- Pescara, 66100 Chieti, Italy.
| | - Emira D'Amico
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology CAST, "G. d' Annunzio" University Chieti- Pescara, 66100 Chieti, Italy
| | - Tea Romasco
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology CAST, "G. d' Annunzio" University Chieti- Pescara, 66100 Chieti, Italy
| | - Maria Laura Gatto
- Department of Science and Engineering of Materials, Environment and Urban Planning, Università Politecnica delle Marche, 60131 Ancona, Italy
| | - Maria Pina Notarangelo
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Carlo Mangano
- Department of Dental Sciences, University Vita Salute San Raffaele, Milan, Italy
| | - Michele Furlani
- Department of Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy
| | - Letizia Penolazzi
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
8
|
Chonanant C, Chancharoen P, Kiatkulanusorn S, Luangpon N, Klarod K, Surakul P, Thamwiriyasati N, Singsanan S, Ngernyuang N. Biocomposite Scaffolds Based on Chitosan Extraction from Shrimp Shell Waste for Cartilage Tissue Engineering Application. ACS OMEGA 2024; 9:39419-39429. [PMID: 39346874 PMCID: PMC11425810 DOI: 10.1021/acsomega.4c02910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/27/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024]
Abstract
Chitosan-based scaffolding possesses unique properties that make it highly suitable for tissue engineering applications. Chitosan is derived from deacetylating chitin, which is particularly abundant in the shells of crustaceans. This study aimed to extract chitosan from shrimp shell waste (Macrobrachium rosenbergii) and produce biocomposite scaffolds using the extracted chitosan for cartilage tissue engineering applications. Chitinous material from shrimp shell waste was deproteinized and deacetylated. The extracted chitosan was characterized and compared to commercial chitosan through various physicochemical analyses. The findings revealed that the extracted chitosan shares similar trends in the Fourier transform infrared spectroscopy spectrum, energy dispersive X-ray mapping, and X-ray diffraction pattern to commercial chitosan. Despite differences in the degree of deacetylation, these results underscore its comparable quality. The extracted chitosan was mixed with agarose, collagen, and gelatin to produce the blending biocomposite AG-CH-COL-GEL scaffold by freeze-drying method. Results showed AG-CH-COL-GEL scaffolds have a 3D interconnected porous structure with pore size 88-278 μm, high water uptake capacity (>90%), and degradation percentages in 21 days between 5.08% and 30.29%. Mechanical compression testing revealed that the elastic modulus of AG-CH-COL-GEL scaffolds ranged from 44.91 to 201.77 KPa. Moreover, AG-CH-COL-GEL scaffolds have shown significant potential in effectively inducing human chondrocyte proliferation and enhancing aggrecan gene expression. In conclusion, AG-CH-COL-GEL scaffolds emerge as promising candidates for cartilage tissue engineering with their optimal physical properties and excellent biocompatibility. This study highlights the potential of using waste-derived chitosan and opens new avenues for sustainable and effective tissue engineering solutions.
Collapse
Affiliation(s)
- Chirapond Chonanant
- Department
of Medical Technology, Faculty of Allied Health Science, Burapha University, Chonburi, 20131, Thailand
| | - Pongrung Chancharoen
- Department
of Medical Sciences, Faculty of Allied Health Sciences, Burapha University, Chonburi 20131, Thailand
| | - Sirirat Kiatkulanusorn
- Department
of Physical Therapy, Faculty of Allied Health Sciences, Burapha University, Chonburi 20131, Thailand
| | - Nongnuch Luangpon
- Department
of Physical Therapy, Faculty of Allied Health Sciences, Burapha University, Chonburi 20131, Thailand
| | - Kultida Klarod
- Department
of Physical Therapy, Faculty of Allied Health Sciences, Burapha University, Chonburi 20131, Thailand
| | - Pornprom Surakul
- Department
of Physical Therapy, Faculty of Allied Health Sciences, Burapha University, Chonburi 20131, Thailand
| | - Niramon Thamwiriyasati
- Department
of Medical Technology, Faculty of Allied Health Science, Burapha University, Chonburi, 20131, Thailand
| | - Sanita Singsanan
- Department
of Medical Technology, Faculty of Allied Health Science, Burapha University, Chonburi, 20131, Thailand
| | - Nipaporn Ngernyuang
- Thammasat
University Research Unit in Biomedical Science, Thammasat University, Pathum
Thani 12120, Thailand
- Chulabhorn
International College of Medicine, Thammasat
University, Pathum
Thani 12120, Thailand
| |
Collapse
|
9
|
Yuan S, Yuan H, Hay DC, Hu H, Wang C. Revolutionizing Drug Discovery: The Impact of Distinct Designs and Biosensor Integration in Microfluidics-Based Organ-on-a-Chip Technology. BIOSENSORS 2024; 14:425. [PMID: 39329800 PMCID: PMC11430660 DOI: 10.3390/bios14090425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
Traditional drug development is a long and expensive process with high rates of failure. This has prompted the pharmaceutical industry to seek more efficient drug development frameworks, driving the emergence of organ-on-a-chip (OOC) based on microfluidic technologies. Unlike traditional animal experiments, OOC systems provide a more accurate simulation of human organ microenvironments and physiological responses, therefore offering a cost-effective and efficient platform for biomedical research, particularly in the development of new medicines. Additionally, OOC systems enable quick and real-time analysis, high-throughput experimentation, and automation. These advantages have shown significant promise in enhancing the drug development process. The success of an OOC system hinges on the integration of specific designs, manufacturing techniques, and biosensors to meet the need for integrated multiparameter datasets. This review focuses on the manufacturing, design, sensing systems, and applications of OOC systems, highlighting their design and sensing capabilities, as well as the technical challenges they currently face.
Collapse
Affiliation(s)
- Sheng Yuan
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
| | - Huipu Yuan
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - David C. Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK;
| | - Huan Hu
- Zhejiang University-University of Illinois Urbana-Champaign Institute (ZJU-UIUC Institute), International Campus, Zhejiang University, Haining 314400, China
| | - Chaochen Wang
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| |
Collapse
|
10
|
Sharma NS, Karan A, Tran HQ, John JV, Andrabi SM, Shatil Shahriar SM, Xie J. Decellularized extracellular matrix-decorated 3D nanofiber scaffolds enhance cellular responses and tissue regeneration. Acta Biomater 2024; 184:81-97. [PMID: 38908416 DOI: 10.1016/j.actbio.2024.06.020] [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: 03/04/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
The use of decellularized extracellular matrix products in tissue regeneration is quite alluring yet practically challenging due to the limitations of its availability, harsh processing techniques, and host rejection. Scaffolds obtained by either incorporating extracellular matrix (ECM) material or coating the surface can resolve these challenges to some extent. However, these scaffolds lack the complex 3D network formed by proteins and growth factors observed in natural ECM. This study introduces an approach utilizing 3D nanofiber scaffolds decorated with dECM to enhance cellular responses and promote tissue regeneration. Notably, the dECM can be customized according to specific cellular requirements, offering a tailored environment for enhanced therapeutic outcomes. Two types of 3D expanded scaffolds, namely radially aligned scaffolds (RAS) and laterally expanded scaffolds (LES) fabricated by the gas-foaming expansion were utilized. To demonstrate the proof-of-concept, human dermal fibroblasts (HDFs) seeded on these scaffolds for up to 8 weeks, resulted in uniform and highly aligned cells which deposited ECM on the scaffolds. These cellular components were then removed from the scaffolds through decellularization (e.g., SDS treatment and freeze-thaw cycles). The dECM-decorated 3D expanded nanofiber scaffolds can direct and support cell alignment and proliferation along the underlying fibers upon recellularization. An in vitro inflammation assay indicates that dECM-decorated LES induces a lower immune response than dECM-decorated RAS. Further, subcutaneous implantation of dECM-decorated RAS and LES shows higher cell infiltration and angiogenesis within 7 and 14 days than RAS and LES without dECM decoration. Taken together, dECM-decorated 3D expanded nanofiber scaffolds hold great potential in tissue regeneration and tissue modeling. STATEMENT OF SIGNIFICANCE: Decellularized ECM scaffolds have attained widespread attention in biomedical applications due to their intricate 3D framework of proteins and growth factors. Mimicking such a complicated architecture is a clinical challenge. In this study, we developed natural ECM-decorated 3D electrospun nanofiber scaffolds with controlled alignments to mimic human tissue. Fibroblasts were cultured on these scaffolds for 8 weeks to deposit natural ECM and decellularized by either freeze-thawing or detergent to obtain decellularized ECM scaffolds. These scaffolds were tested in both in-vitro and in-vivo conditions. They displayed higher cellular attributes with lower immune response making them a good grafting tool in tissue regeneration.
Collapse
Affiliation(s)
- Navatha Shree Sharma
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center Omaha, NE 68198, United States
| | - Anik Karan
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center Omaha, NE 68198, United States
| | - Huy Quang Tran
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center Omaha, NE 68198, United States
| | - Johnson V John
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, United States
| | - Syed Muntazir Andrabi
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center Omaha, NE 68198, United States
| | - S M Shatil Shahriar
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center Omaha, NE 68198, United States
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center Omaha, NE 68198, United States; Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, United States.
| |
Collapse
|
11
|
Lai X, Huang J, Huang S, Wang J, Zheng Y, Luo Y, Tang L, Gao B, Tang Y. Antibacterial and Osteogenic Dual-Functional Micronano Composite Scaffold Fabricated via Melt Electrowriting and Solution Electrospinning for Bone Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37707-37721. [PMID: 39001812 DOI: 10.1021/acsami.4c07400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/15/2024]
Abstract
The utilization of micronano composite scaffolds has been extensively demonstrated to confer the superior advantages in bone repair compared to single nano- or micron-sized scaffolds. Nevertheless, the enhancement of bioactivities within these composite scaffolds remains challenging. In this study, we propose a novel approach to combine melt electrowriting (MEW) and solution electrospinning (SES) techniques for the fabrication of a composite scaffold incorporating hydroxyapatite (HAP), an osteogenic component, and roxithromycin (ROX), an antibacterial active component. Scanning electron microscopy (SEM) and Fourier-transform infrared spectroscopy (FTIR) confirmed the hierarchical architecture of the nanofiber-microgrid within the scaffold, as well as the successful loading of HAP and ROX. The incorporation of HAP enhanced the water absorption capacity of the composite scaffold, thus promoting cell adhesion and proliferation, as well as osteogenic differentiation. Furthermore, ROX resulted in effective antibacterial capability without any observable cytotoxicity. Finally, the scaffolds were applied to a rat calvarial defect model, and the results demonstrated that the 20% HAP group exhibited superior new bone formation without causing adverse reactions. Therefore, our findings present a promising strategy for designing and fabricating bioactive scaffolds for bone regeneration.
Collapse
Affiliation(s)
- Xiangjie Lai
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Jun Huang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, China
| | - Shunfen Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, China
| | - Jiyuan Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yongsheng Zheng
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, China
- Department of Orthopedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yuli Luo
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Linjun Tang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, China
| | - Yadong Tang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
12
|
Chen X, Zhou Z, Yang M, Zhu S, Zhu W, Sun J, Yu M, He J, Zuo Y, Wang W, He N, Han X, Liu H. A biocompatible pea protein isolate-derived bioink for 3D bioprinting and tissue engineering. J Mater Chem B 2024; 12:6716-6723. [PMID: 38899871 DOI: 10.1039/d4tb00781f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Three-dimensional bioprinting is a potent biofabrication technique in tissue engineering but is limited by inadequate bioink availability. Plant-derived proteins are increasingly recognized as highly promising yet underutilized materials for biomedical product development and hold potential for use in bioink formulations. Herein, we report the development of a biocompatible plant protein bioink from pea protein isolate. Through pH shifting, ethanol precipitation, and lyophilization, the pea protein isolate (PPI) transformed from an insoluble to a soluble form. Next, it was modified with glycidyl methacrylate to obtain methacrylate-modified PPI (PPIGMA), which is photocurable and was used as the precursor of bioink. The mechanical and microstructural studies of the hydrogel containing 16% PPIGMA revealed a suitable compress modulus and a porous network with a pore size over 100 μm, which can facilitate nutrient and waste transportation. The PPIGMA bioink exhibited good 3D bioprinting performance in creating complex patterns and good biocompatibility as plenty of viable cells were observed in the printed samples after 3 days of incubation in the cell culture medium. No immunogenicity of the PPIGMA bioink was identified as no inflammation was observed for 4 weeks after implantation in Sprague Dawley rats. Compared with methacrylate-modified gelatin, the PPIGMA bioink significantly enhanced cartilage regeneration in vitro and in vivo, suggesting that it can be used in tissue engineering applications. In summary, the PPIGMA bioink can be potentially used for tissue engineering applications.
Collapse
Affiliation(s)
- Xin Chen
- College of Material Science and Engineering, Hunan University, Changsha 410082, China.
| | - Zheng Zhou
- College of Biology, Hunan University, Changsha 410082, China.
| | - Mengni Yang
- College of Material Science and Engineering, Hunan University, Changsha 410082, China.
| | - Shuai Zhu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China.
| | - Wenxiang Zhu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China.
| | - Jingjing Sun
- College of Biology, Hunan University, Changsha 410082, China.
| | - Mengyi Yu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China.
| | - Jiaqian He
- College of Biology, Hunan University, Changsha 410082, China.
| | - You Zuo
- College of Biology, Hunan University, Changsha 410082, China.
| | - Wenxin Wang
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha 410082, China
| | - Ning He
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha 410082, China
| | - Xiaoxiao Han
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha 410082, China
| | - Hairong Liu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China.
| |
Collapse
|
13
|
Komatsu D, Cabrera ARE, Quevedo BV, Asami J, Cristina Motta A, de Moraes SC, Duarte MAT, Hausen MDA, Aparecida de Rezende Duek E. Meniscal repair with additive manufacture of bioresorbable polymer: From physicochemical characterization to implantation of 3D printed poly (L-co-D, L lactide-co-trimethylene carbonate) with autologous stem cells in rabbits. J Biomater Appl 2024; 39:66-79. [PMID: 38646887 DOI: 10.1177/08853282241248517] [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] [Indexed: 04/23/2024]
Abstract
Three-dimensional (3D) structures are actually the state-of-the-art technique to create porous scaffolds for tissue engineering. Since regeneration in cartilage tissue is limited due to intrinsic cellular properties this study aims to develop and characterize three-dimensional porous scaffolds of poly (L-co-D, L lactide-co-trimethylene carbonate), PLDLA-TMC, obtained by 3D fiber deposition technique. The PLDLA-TMC terpolymer scaffolds (70:30), were obtained and characterized by scanning electron microscopy, gel permeation chromatography, differential scanning calorimetry, thermal gravimetric analysis, compression mechanical testing and study on in vitro degradation, which showed its amorphous characteristics, cylindrical geometry, and interconnected pores. The in vitro degradation study showed significant loss of mechanical properties compatible with a decrease in molar mass, accompanied by changes in morphology. The histocompatibility association of mesenchymal stem cells from rabbit's bone marrow, and PLDLA-TMC scaffolds, were evaluated in the meniscus regeneration, proving the potential of cell culture at in vivo tissue regeneration. Nine New Zealand rabbits underwent total medial meniscectomy, yielding three treatments: implantation of the seeded PLDLA-TMC scaffold, implantation of the unseeded PLDLA-TMC and negative control (defect without any implant). After 24 weeks, the results revealed the presence of fibrocartilage in the animals treated with polymer. However, the regeneration obtained with the seeded PLDLA-TMC scaffolds with mesenchymal stem cells had become intimal to mature fibrocartilaginous tissue of normal meniscus both macroscopically and histologically. This study demonstrated the effectiveness of the PLDLA-TMC scaffold in meniscus regeneration and the potential of mesenchymal stem cells in tissue engineering, without the use of growth factors. It is concluded that bioresorbable polymers represent a promising alternative for tissue regeneration.
Collapse
Affiliation(s)
- Daniel Komatsu
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
| | | | - Bruna Vanessa Quevedo
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
- Post-Graduation Program in Materials Sciences (PPGCM), Federal University of São Carlos (UFSCar), Sorocaba, Brazil
| | - Jessica Asami
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
- Post-Graduation Program of School of Mechanical Engineering (FEM), University of Campinas (UNICAMP), Campinas, Brazil
| | - Adriana Cristina Motta
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
| | | | | | - Moema de Alencar Hausen
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
- Post-Graduation Program of Biomaterials and Regenerative Medicine (PPGBMR), Surgery Department, PUC-SP, Sorocaba, Brazil
| | - Eliana Aparecida de Rezende Duek
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
- Post-Graduation Program of School of Mechanical Engineering (FEM), University of Campinas (UNICAMP), Campinas, Brazil
- Post-Graduation Program in Materials Sciences (PPGCM), Federal University of São Carlos (UFSCar), Sorocaba, Brazil
- Post-Graduation Program of Biomaterials and Regenerative Medicine (PPGBMR), Surgery Department, PUC-SP, Sorocaba, Brazil
| |
Collapse
|
14
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [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: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
15
|
Bae KB, Kim HM, Son JW, Ryu JY, Hwang YC, Koh JT, Oh WM, Park C, Lee BN. Effect of 3D-printed polycaprolactone/osteolectin scaffolds on the odontogenic differentiation of human dental pulp cells. Biomed Mater 2024; 19:045027. [PMID: 38740059 DOI: 10.1088/1748-605x/ad4ad9] [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: 01/25/2024] [Accepted: 05/13/2024] [Indexed: 05/16/2024]
Abstract
Cell-based tissue engineering often requires the use of scaffolds to provide a three-dimensional (3D) framework for cell proliferation and tissue formation. Polycaprolactone (PCL), a type of polymer, has good printability, favorable surface modifiability, adaptability, and biodegradability. However, its large-scale applicability is hindered by its hydrophobic nature, which affects biological properties. Composite materials can be created by adding bioactive materials to the polymer to improve the properties of PCL scaffolds. Osteolectin is an odontogenic factor that promotes the maintenance of the adult skeleton by promoting the differentiation of LepR+ cells into osteoblasts. Therefore, the aim of this study was to evaluate whether 3D-printed PCL/osteolectin scaffolds supply a suitable microenvironment for the odontogenic differentiation of human dental pulp cells (hDPCs). The hDPCs were cultured on 3D-printed PCL scaffolds with or without pores. Cell attachment and cell proliferation were evaluated using EZ-Cytox. The odontogenic differentiation of hDPCs was evaluated by alizarin red S staining and alkaline phosphatase assays. Western blot was used to evaluate the expression of the proteins DSPP and DMP-Results: The attachment of hDPCs to PCL scaffolds with pores was significantly higher than to PCL scaffolds without pores. The odontogenic differentiation of hDPCs was induced more in PCL/osteolectin scaffolds than in PCL scaffolds, but there was no statistically significant difference. 3D-printed PCL scaffolds with pores are suitable for the growth of hDPCs, and the PCL/osteolectin scaffolds can provide a more favorable microenvironment for the odontogenic differentiation of hDPCs.
Collapse
Affiliation(s)
- Kkot-Byeol Bae
- Department of Conservative Dentistry, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Hae-Mi Kim
- Private practice, Local Dental Clinic, Seoul, Republic of Korea
| | - Ji-Won Son
- Researcher, Department of Conservative Dentistry, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Jae-Young Ryu
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Yun-Chan Hwang
- Department of Conservative Dentistry, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Jeong-Tae Koh
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Won-Mann Oh
- Department of Conservative Dentistry, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Chan Park
- Department of Prosthodontics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Bin-Na Lee
- Department of Conservative Dentistry, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
16
|
Karaca I, Aldemir Dikici B. Quantitative Evaluation of the Pore and Window Sizes of Tissue Engineering Scaffolds on Scanning Electron Microscope Images Using Deep Learning. ACS OMEGA 2024; 9:24695-24706. [PMID: 38882138 PMCID: PMC11170757 DOI: 10.1021/acsomega.4c01234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 06/18/2024]
Abstract
The morphological characteristics of tissue engineering scaffolds, such as pore and window diameters, are crucial, as they directly impact cell-material interactions, attachment, spreading, infiltration of the cells, degradation rate and the mechanical properties of the scaffolds. Scanning electron microscopy (SEM) is one of the most commonly used techniques for characterizing the microarchitecture of tissue engineering scaffolds due to its advantages, such as being easily accessible and having a short examination time. However, SEM images provide qualitative data that need to be manually measured using software such as ImageJ to quantify the morphological features of the scaffolds. As it is not practical to measure each pore/window in the SEM images as it requires extensive time and effort, only the number of pores/windows is measured and assumed to represent the whole sample, which may cause user bias. Additionally, depending on the number of samples and groups, a study may require measuring thousands of samples and the human error rate may increase. To overcome such problems, in this study, a deep learning model (Pore D2) was developed to quantify the morphological features (such as the pore size and window size) of the open-porous scaffolds automatically for the first time. The developed algorithm was tested on emulsion-templated scaffolds fabricated under different fabrication conditions, such as changing mixing speed, temperature, and surfactant concentration, which resulted in scaffolds with various morphologies. Along with the developed model, blind manual measurements were taken, and the results showed that the developed tool is capable of quantifying pore and window sizes with a high accuracy. Quantifying the morphological features of scaffolds fabricated under different circumstances and controlling these features enable us to engineer tissue engineering scaffolds precisely for specific applications. Pore D2, an open-source software, is available for everyone at the following link: https://github.com/ilaydakaraca/PoreD2.
Collapse
Affiliation(s)
- Ilayda Karaca
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Betül Aldemir Dikici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| |
Collapse
|
17
|
Yao Y, Chen K, Pan Q, Gao H, Su W, Zheng S, Dong W, Qian D. Redifferentiation of genetically modified dedifferentiated chondrocytes in a microcavitary hydrogel. Biotechnol Lett 2024; 46:483-495. [PMID: 38523201 DOI: 10.1007/s10529-024-03475-2] [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: 10/10/2023] [Revised: 01/11/2024] [Accepted: 02/15/2024] [Indexed: 03/26/2024]
Abstract
OBJECTIVES We genetically modified dedifferentiated chondrocytes (DCs) using lentiviral vectors and adenoviral vectors encoding TGF-β3 (referred to as transgenic groups below) and encapsulated these DCs in the microcavitary hydrogel and investigated the combinational effect on redifferentiation of the genetically manipulated DCs. RESULTS The Cell Counting Kit-8 data indicated that both transgenic groups exhibited significantly higher cell viability in the first week but inferior cell viability in the subsequent timepoints compared with those of the control group. Real-time polymerase chain reaction and western blot analysis results demonstrated that both transgenic groups had a better effect on redifferentiation to some extent, as evidenced by higher expression levels of chondrogenic genes, suggesting the validity of combination with transgenic DCs and the microcavitary hydrogel on redifferentiation. Although transgenic DCs with adenoviral vectors presented a superior extent of redifferentiation, they also expressed greater levels of the hypertrophic gene type X collagen. It is still worth further exploring how to deliver TGF-β3 more efficiently and optimizing the appropriate parameters, including concentration and duration. CONCLUSIONS The results demonstrated the better redifferentiation effect of DCs with the combinational use of transgenic TGF-β3 and a microcavitary alginate hydrogel and implied that DCs would be alternative seed cells for cartilage tissue engineering due to their easily achieved sufficient cell amounts through multiple passages and great potential to redifferentiate to produce cartilaginous extracellular matrix.
Collapse
Affiliation(s)
- Yongchang Yao
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
| | - Ke Chen
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- Emergency Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
| | - Qian Pan
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Hui Gao
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Weixian Su
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Shicong Zheng
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Weiqiang Dong
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Dongyang Qian
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| |
Collapse
|
18
|
Chen Y, Li Y, Zhu W, Liu Q. Biomimetic gradient scaffolds for the tissue engineering and regeneration of rotator cuff enthesis. Biofabrication 2024; 16:032005. [PMID: 38697099 DOI: 10.1088/1758-5090/ad467d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/02/2024] [Indexed: 05/04/2024]
Abstract
Rotator cuff tear is one of the most common musculoskeletal disorders, which often results in recurrent shoulder pain and limited movement. Enthesis is a structurally complex and functionally critical interface connecting tendon and bone that plays an essential role in maintaining integrity of the shoulder joint. Despite the availability of advanced surgical procedures for rotator cuff repair, there is a high rate of failure following surgery due to suboptimal enthesis healing and regeneration. Novel strategies based on tissue engineering are gaining popularity in improving tendon-bone interface (TBI) regeneration. Through incorporating physical and biochemical cues into scaffold design which mimics the structure and composition of native enthesis is advantageous to guide specific differentiation of seeding cells and facilitate the formation of functional tissues. In this review, we summarize the current state of research in enthesis tissue engineering highlighting the development and application of biomimetic scaffolds that replicate the gradient TBI. We also discuss the latest techniques for fabricating potential translatable scaffolds such as 3D bioprinting and microfluidic device. While preclinical studies have demonstrated encouraging results of biomimetic gradient scaffolds, the translation of these findings into clinical applications necessitates a comprehensive understanding of their safety and long-term efficacy.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yexin Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Qian Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
19
|
Kolodkin-Gal I, Dash O, Rak R. Probiotic cultivated meat: bacterial-based scaffolds and products to improve cultivated meat. Trends Biotechnol 2024; 42:269-281. [PMID: 37805297 DOI: 10.1016/j.tibtech.2023.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 10/09/2023]
Abstract
Cultivated meat is emerging to replace traditional livestock industries, which have ecological costs, including land and water overuse and considerable carbon emissions. During cultivated meat production, mammalian cells can increase their numbers dramatically through self-renewal/proliferation and transform into mature cells, such as muscle or fat cells, through maturation/differentiation. Here, we address opportunities for introducing probiotic bacteria into the cultivated meat industry, including using them to produce renewable antimicrobials and scaffolding materials. We also offer solutions to challenges, including the growth of bacteria and mammalian cells, the effect of probiotic bacteria on production costs, and the effect of bacteria and their products on texture and taste. Our summary provides a promising framework for applying microbial composites in the cultivated meat industry.
Collapse
Affiliation(s)
- Ilana Kolodkin-Gal
- Scojen Institute for Synthetic Biology, Reichman University, Herzliya, Israel.
| | - Orit Dash
- Department of Animal Sciences, Faculty of Agriculture and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel; Institute of Animal Science, ARO, The Volcani Center, Rishon LeZion, Israel
| | - Roni Rak
- Institute of Animal Science, ARO, The Volcani Center, Rishon LeZion, Israel.
| |
Collapse
|
20
|
Trifonov A, Shehzad A, Mukasheva F, Moazzam M, Akilbekova D. Reasoning on Pore Terminology in 3D Bioprinting. Gels 2024; 10:153. [PMID: 38391483 PMCID: PMC10887720 DOI: 10.3390/gels10020153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
Terminology is pivotal for facilitating clear communication and minimizing ambiguity, especially in specialized fields such as chemistry. In materials science, a subset of chemistry, the term "pore" is traditionally linked to the International Union of Pure and Applied Chemistry (IUPAC) nomenclature, which categorizes pores into "micro", "meso", and "macro" based on size. However, applying this terminology in closely-related areas, such as 3D bioprinting, often leads to confusion owing to the lack of consensus on specific definitions and classifications tailored to each field. This review article critically examines the current use of pore terminology in the context of 3D bioprinting, highlighting the need for reassessment to avoid potential misunderstandings. We propose an alternative classification that aligns more closely with the specific requirements of bioprinting, suggesting a tentative size-based division of interconnected pores into 'parvo'-(d < 25 µm), 'medio'-(25 < d < 100 µm), and 'magno'-(d > 100 µm) pores, relying on the current understanding of the pore size role in tissue formation. The introduction of field-specific terminology for pore sizes in 3D bioprinting is essential to enhance the clarity and precision of research communication. This represents a step toward a more cohesive and specialized lexicon that aligns with the unique aspects of bioprinting and tissue engineering.
Collapse
Affiliation(s)
- Alexander Trifonov
- Department of Chemical and Materials Engineering, School of Engineering, Nazarbayev University, Astana 010000, Kazakhstan
| | - Ahmer Shehzad
- Department of Chemical and Materials Engineering, School of Engineering, Nazarbayev University, Astana 010000, Kazakhstan
| | - Fariza Mukasheva
- Department of Chemical and Materials Engineering, School of Engineering, Nazarbayev University, Astana 010000, Kazakhstan
| | - Muhammad Moazzam
- Department of Chemical and Materials Engineering, School of Engineering, Nazarbayev University, Astana 010000, Kazakhstan
| | - Dana Akilbekova
- Department of Chemical and Materials Engineering, School of Engineering, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
21
|
Pfaff BN, Flanagan CC, Griffin DR. Microporous Annealed Particle (MAP) Scaffold Pore Size Influences Mesenchymal Stem Cell Metabolism and Proliferation Without Changing CD73, CD90, and CD105 Expression Over Two Weeks. Adv Biol (Weinh) 2024; 8:e2300482. [PMID: 37955859 PMCID: PMC10922193 DOI: 10.1002/adbi.202300482] [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: 09/07/2023] [Revised: 10/25/2023] [Indexed: 11/14/2023]
Abstract
Scaffold pore architecture is shown to influence stem cell fate through various avenues. It is demonstrated that microporous annealed particle (MAP) microgel diameter can be tuned to control scaffold pore size and, in turn, modulate mesenchymal stem cell (MSC) survivability, proliferation, metabolism, and migration, thereby enhancing bioactivity and guiding future applications of MAP for regenerative medicine.
Collapse
Affiliation(s)
- Blaise N Pfaff
- University of Virginia, 415 Lane Road, Charlottesville, VA, 22903, USA
| | - Clare C Flanagan
- University of Virginia, 415 Lane Road, Charlottesville, VA, 22903, USA
| | - Donald R Griffin
- University of Virginia, 415 Lane Road, Charlottesville, VA, 22903, USA
- Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, VA, 22903-3390, USA
| |
Collapse
|
22
|
Ozkendir O, Karaca I, Cullu S, Erdoğan OC, Yaşar HN, Dikici S, Owen R, Aldemir Dikici B. Engineering periodontal tissue interfaces using multiphasic scaffolds and membranes for guided bone and tissue regeneration. BIOMATERIALS ADVANCES 2024; 157:213732. [PMID: 38134730 DOI: 10.1016/j.bioadv.2023.213732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Periodontal diseases are one of the greatest healthcare burdens worldwide. The periodontal tissue compartment is an anatomical tissue interface formed from the periodontal ligament, gingiva, cementum, and bone. This multifaceted composition makes tissue engineering strategies challenging to develop due to the interface of hard and soft tissues requiring multiphase scaffolds to recreate the native tissue architecture. Multilayer constructs can better mimic tissue interfaces due to the individually tuneable layers. They have different characteristics in each layer, with modulation of mechanical properties, material type, porosity, pore size, morphology, degradation properties, and drug-releasing profile all possible. The greatest challenge of multilayer constructs is to mechanically integrate consecutive layers to avoid delamination, especially when using multiple manufacturing processes. Here, we review the development of multilayer scaffolds that aim to recapitulate native periodontal tissue interfaces in terms of physical, chemical, and biological characteristics. Important properties of multiphasic biodegradable scaffolds are highlighted and summarised, with design requirements, biomaterials, and fabrication methods, as well as post-treatment and drug/growth factor incorporation discussed.
Collapse
Affiliation(s)
- Ozgu Ozkendir
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Ilayda Karaca
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Selin Cullu
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Oğul Can Erdoğan
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Hüsniye Nur Yaşar
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Serkan Dikici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Robert Owen
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Betül Aldemir Dikici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey.
| |
Collapse
|
23
|
Kohoolat G, Alizadeh P, Motesadi Zarandi F, Rezaeipour Y. A ternary composite hydrogel based on sodium alginate, carboxymethyl cellulose and copper-doped 58S bioactive glass promotes cutaneous wound healing in vitro and in vivo. Int J Biol Macromol 2024; 259:129260. [PMID: 38199544 DOI: 10.1016/j.ijbiomac.2024.129260] [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: 08/14/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
Hydrogels offer a novel approach to wound repair. In this study, we synthesized a ternary composite using sodium alginate (SA), carboxymethyl cellulose (CMC) and copper-doped 58S bioactive glass (BG). According to our mechanical testing results, the composite made of 7 wt% CMC and 7 wt% BG (SA-7CMC-7BG) showed optimal properties. In addition, our in vitro studies revealed the biocompatibility and bioactivity of SA-7CMC-7BG, with a negative zeta potential of -31.7 mV. Scanning electron microscope (SEM) images showed 273-μm-diameter pores, cell adhesion, and anchoring. The SA-7CMC-7BG closed 90.4 % of the mechanical scratch after 2 days. An in vivo wound model using Wistar rats showed that SA-7CMC-7BG promoted wound healing, with 85.57 % of the wounds healed after 14 days. Treatment with the SA-7CMC-7BG hydrogel caused a 1.6-, 65-, and 1.87-fold increase in transforming growth factor beta (TGF-β), Col I, and vascular endothelial growth factor (VEGF) expression, respectively that prevents fibrosis and promotes angiogenesis. Furthermore, interleukin 1β (IL-1β) expression was downregulated by 1.61-fold, indicating an anti-inflammatory effect of SA-7CMC-7BG. We also observed an increase in epidermal thickness, the number of fibroblast cells, and collagen deposition, which represent complementary pathology results confirming the effectiveness of the SA-7CMC-7BG hydrogel in cutaneous wound healing.
Collapse
Affiliation(s)
- Ghazaleh Kohoolat
- Department of Materials Science & Engineering, Faculty of Engineering & Technology, Tarbiat Modares University, P. O. Box: 14115-143, Tehran, Iran
| | - Parvin Alizadeh
- Department of Materials Science & Engineering, Faculty of Engineering & Technology, Tarbiat Modares University, P. O. Box: 14115-143, Tehran, Iran.
| | - Fatemeh Motesadi Zarandi
- Department of Materials Science & Engineering, Faculty of Engineering & Technology, Tarbiat Modares University, P. O. Box: 14115-143, Tehran, Iran
| | - Yashar Rezaeipour
- Department of Materials Science & Engineering, Faculty of Engineering & Technology, Tarbiat Modares University, P. O. Box: 14115-143, Tehran, Iran
| |
Collapse
|
24
|
Galindo JM, San-Millán MI, Castillo-Sarmiento CA, Ballesteros-Yáñez I, Vázquez E, Merino S, Herrero MA. Optimization of 3D Synthetic Scaffolds for Neuronal Tissue Engineering Applications. Chemistry 2024; 30:e202302481. [PMID: 37823243 DOI: 10.1002/chem.202302481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023]
Abstract
The increasing prevalence of neurodegenerative diseases has spurred researchers to develop advanced 3D models that accurately mimic neural tissues. Hydrogels stand out as ideal candidates as their properties closely resemble those of the extracellular matrix. A critical challenge in this regard is to comprehend the influence of the scaffold's mechanical properties on cell growth and differentiation, thus enabling targeted modifications. In light of this, a synthesis and comprehensive analysis of acrylamide-based hydrogels incorporating a peptide has been conducted. Adequate cell adhesion and development is achieved due to their bioactive nature and specific interactions with cellular receptors. The integration of a precisely controlled physicochemical hydrogel matrix and inclusion of the arginine-glycine-aspartic acid peptide sequence has endowed this system with an optimal structure, thus providing a unique ability to interact effectively with biomolecules. The analysis fully examined essential properties governing cell behavior, including pore size, mechanical characteristics, and swelling ability. Cell-viability experiments were performed to assess the hydrogel's biocompatibility, while the incorporation of grow factors aimed to promote the differentiation of neuroblastoma cells. The results underscore the hydrogel's ability to stimulate cell viability and differentiation in the presence of the peptide within the matrix.
Collapse
Affiliation(s)
- Josué M Galindo
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Ms Irene San-Millán
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | | | | | - Ester Vázquez
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Sonia Merino
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - M Antonia Herrero
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| |
Collapse
|
25
|
Huang YH, Chen HA, Chen CH, Liao HT, Kuo CY, Chen JP. Injectable gelatin/glucosamine cryogel microbeads as scaffolds for chondrocyte delivery in cartilage tissue engineering. Int J Biol Macromol 2023; 253:126528. [PMID: 37633562 DOI: 10.1016/j.ijbiomac.2023.126528] [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: 07/05/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
In this study, we fabricate squeezable cryogel microbeads as injectable scaffolds for minimum invasive delivery of chondrocytes for cartilage tissue engineering applications. The microbeads with different glucosamine concentrations were prepared by combining the water-in-oil emulsion and cryogelation through crosslinking of gelatin with glutaraldehyde in the presence of glucosamine. The physicochemical characterization results show the successful preparation of cryogel microbeads with uniform shape and size, high porosity, large pore size, high water uptake capacity, and good injectability. In vitro analysis indicates proliferation, migration, and differentiated phenotype of rabbit chondrocytes in the cryogel scaffolds. The seeded chondrocytes in the cryogel scaffold can be delivered by injecting through an 18G needle to fully retain the cell viability. Furthermore, the incorporation of glucosamine in the cryogel promoted the differentiated phenotype of chondrocytes in a dose-dependent manner, from cartilage-specific gene expression and protein production. The in vivo study by injecting the cryogel microbeads into the subcutaneous pockets of nude mice indicates good retention ability as well as good biocompatibility and suitable biodegradability of the cryogel scaffold. Furthermore, the injected chondrocyte/cryogel microbead constructs can form ectopic functional neocartilage tissues following subcutaneous implantation in 21 days, as evidenced by histological and immunohistochemical analysis.
Collapse
Affiliation(s)
- Yen-Hsiang Huang
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Huai-An Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Chih-Hao Chen
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Keelung, Keelung 20401, Taiwan; Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan
| | - Han-Tsung Liao
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan
| | - Chang-Yi Kuo
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan; Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33305, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
26
|
Weekes A, Wehr G, Pinto N, Jenkins J, Li Z, Meinert C, Klein TJ. Highly compliant biomimetic scaffolds for small diameter tissue-engineered vascular grafts (TEVGs) produced via melt electrowriting (MEW). Biofabrication 2023; 16:015017. [PMID: 37992322 DOI: 10.1088/1758-5090/ad0ee1] [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: 06/05/2023] [Accepted: 11/22/2023] [Indexed: 11/24/2023]
Abstract
Biofabrication approaches toward the development of tissue-engineered vascular grafts (TEVGs) have been widely investigated. However, successful translation has been limited to large diameter applications, with small diameter grafts frequently failing due to poor mechanical performance, in particular mismatched radial compliance. Herein, melt electrowriting (MEW) of poly(ϵ-caprolactone) has enabled the manufacture of highly porous, biocompatible microfibre scaffolds with physiological anisotropic mechanical properties, as substrates for the biofabrication of small diameter TEVGs. Highly reproducible scaffolds with internal diameter of 4.0 mm were designed with 500 and 250µm pore sizes, demonstrating minimal deviation of less than 4% from the intended architecture, with consistent fibre diameter of 15 ± 2µm across groups. Scaffolds were designed with straight or sinusoidal circumferential microfibre architecture respectively, to investigate the influence of biomimetic fibre straightening on radial compliance. The results demonstrate that scaffolds with wave-like circumferential microfibre laydown patterns mimicking the architectural arrangement of collagen fibres in arteries, exhibit physiological compliance (12.9 ± 0.6% per 100 mmHg), while equivalent control geometries with straight fibres exhibit significantly reduced compliance (5.5 ± 0.1% per 100 mmHg). Further mechanical characterisation revealed the sinusoidal scaffolds designed with 250µm pores exhibited physiologically relevant burst pressures of 1078 ± 236 mmHg, compared to 631 ± 105 mmHg for corresponding 500µm controls. Similar trends were observed for strength and failure, indicating enhanced mechanical performance of scaffolds with reduced pore spacing. Preliminaryin vitroculture of human mesenchymal stem cells validated the MEW scaffolds as suitable substrates for cellular growth and proliferation, with high cell viability (>90%) and coverage (>85%), with subsequent seeding of vascular endothelial cells indicating successful attachment and preliminary endothelialisation of tissue-cultured constructs. These findings support further investigation into long-term tissue culture methodologies for enhanced production of vascular extracellular matrix components, toward the development of the next generation of small diameter TEVGs.
Collapse
Affiliation(s)
- Angus Weekes
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
| | - Gabrielle Wehr
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
| | - Nigel Pinto
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
- Department of Vascular Surgery, The Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Jason Jenkins
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
- Department of Vascular Surgery, The Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Zhiyong Li
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Christoph Meinert
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
| | - Travis J Klein
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| |
Collapse
|
27
|
Cho EH, Kim YS, Kim YR, Kang JH, Park SW, Lim HP, Yun KD, Jang WH, Koh JT, Park C, Lee BN. A leptin-loaded poly- ϵ-caprolactone 3D printing scaffold for odontoblastic differentiation in human dental pulp cells. Biomed Mater 2023; 19:015009. [PMID: 37972541 DOI: 10.1088/1748-605x/ad0d84] [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: 07/06/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023]
Abstract
This study investigated the effects on odontoblast differentiation of a 3D-printed poly-ϵ-caprolactone (PCL) scaffold that incorporated leptin. Material extrusion-type 3D printing with a 43 000-molecular weight PCL material was used to fabricate a PCL scaffold with a 6 mm diameter, 1 mm height, and 270-340 µm pore size. The experimental groups were PCL scaffolds (control group), PCL scaffolds with aminated surfaces (group A), and PCL scaffolds with leptin on the aminated surface (group L). The aminated surface was treated with 1,6-hexanediamine and verified by ninhydrin analysis. Leptin loading was performed using Traut's reagent and 4-(N-Maleimidomethyl)cyclohexane-1-carboxylic acid 3-sulfo-N-hydroxysuccinimide ester sodium salt (Sulfo-SMCC). Groups A and L showed significantly higher surface wettability, pulp cell adhesion, and proliferation than the control group. Group L exhibited increased alkaline phosphatase, calcification deposits, and mRNA and protein expression of dentin sialophosphoprotein and dentin matrix acidic phosphoprotein 1 compared with the control group. In this study, a 3D-printed PCL scaffold containing leptin was enhanced odontoblast differentiation and dental pulp cells adhesion and proliferation.
Collapse
Affiliation(s)
- Eun-Hyo Cho
- School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Ye-Seul Kim
- Department of Prosthodontics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Young-Ran Kim
- Department of Prosthodontics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Jin-Ho Kang
- Department of Prosthodontics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Sang-Won Park
- Department of Prosthodontics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Hyun-Pil Lim
- Department of Prosthodontics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Kwi-Dug Yun
- Department of Prosthodontics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Woo-Hyung Jang
- Department of Prosthodontics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Jeong-Tea Koh
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Chan Park
- Department of Prosthodontics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Bin-Na Lee
- Department of Conservative Dentistry, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
28
|
Abdulaziz D, Anastasiou AD, Panagiotopoulou V, Raif EM, Giannoudis PV, Jha A. Physiologically engineered porous titanium/brushite scaffolds for critical-size bone defects: A design and manufacturing study. J Mech Behav Biomed Mater 2023; 148:106223. [PMID: 37976684 DOI: 10.1016/j.jmbbm.2023.106223] [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: 09/13/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Repairing critical-size bone defects still represents a critical clinical challenge in the field of trauma surgery. This study focuses on a physiological design and manufacturing of porous composite scaffold (titanium Ti with 10 % mole iron doped brushite DCPD-Fe3+) which can mimic the biomechanical properties of natural cortical bone, specifically for the purpose of repairing critical-size defects. To achieve this, the principle of design of experiments (DOE) was applied for investigating the impact of sintering temperature, mineral ratio, and volume fraction of porosity on the mechanical properties of the fabricated scaffolds. The fabricated scaffolds had open porosity up to 60 %, with pore size approximately between 100 μm and 850 μm. The stiffness of the porous composite scaffolds varied between 3.30 GPa and 20.50 GPa, while the compressive strength ranged from approximately 130 MPa-165 MPa at sintering temperatures equal to or exceeding 1000 °C. Scaffolds with higher porosity and mineral content demonstrated lower stiffness values, resembling natural bone. Numerical simulation was employed by Ansys Workbench to investigate the stress and strain distribution of a critical size defect in mid-shaft femur which was designed to be replaced with the fabricated scaffold. The fabricated scaffolds showed flexible biomechanical behaviour at the bone/scaffold interface, generating lower stress levels and indicating a better match with the femoral shaft stiffness. The experimental and numerical findings demonstrated promising applications for manufacturing a patient-specific bone scaffold for critical and potentially large defects for reducing stress shielding and minimizing non-union risk.
Collapse
Affiliation(s)
- Dina Abdulaziz
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, UK.
| | - Antonios D Anastasiou
- Department of Chemical Engineering, University of Manchester, Manchester, M1 3AL, UK
| | | | - El Mostafa Raif
- Faculty of Medicine and Health, School of Dentistry, University of Leeds, Leeds, LS2 9JT, UK
| | - Peter V Giannoudis
- Academic Department of Trauma and Orthopaedic Surgery, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Animesh Jha
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
29
|
Wang X, Li Z, Liu J, Wang C, Bai H, Zhu X, Wang H, Wang Z, Liu H, Wang J. 3D-printed PCL scaffolds with anatomy-inspired bionic stratified structures for the treatment of growth plate injuries. Mater Today Bio 2023; 23:100833. [PMID: 37920293 PMCID: PMC10618519 DOI: 10.1016/j.mtbio.2023.100833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/27/2023] [Accepted: 10/14/2023] [Indexed: 11/04/2023] Open
Abstract
The growth plate is a cartilaginous tissue with three distinct zones. Resident chondrocytes are highly organized in a columnar structure, which is critical for the longitudinal growth of immature long bones. Once injured, the growth plate may potentially be replaced by bony bar formation and, consequently, cause limb abnormalities in children. It is well-known that the essential step in growth plate repair is the remolding of the organized structure of chondrocytes. To achieve this, we prepared an anatomy-inspired bionic Poly(ε-caprolactone) (PCL) scaffold with a stratified structure using three-dimensional (3D) printing technology. The bionic scaffold is engineered by surface modification of NaOH and collagen Ⅰ (COL Ⅰ) to promote cell adhesion. Moreover, chondrocytes and bone marrow mesenchymal stem cells (BMSCs) are loaded in the most suitable ratio of 1:3 for growth plate reconstruction. Based on the anatomical structure of the growth plate, the bionic scaffold is designed to have three regions, which are the small-, medium-, and large-pore-size regions. These pore sizes are used to induce BMSCs to differentiate into similar structures such as the growth plate. Remarkably, the X-ray and histological results also demonstrate that the cell-loaded stratified scaffold can successfully rebuild the structure of the growth plate and reduce limb abnormalities, including limb length discrepancies and angular deformities in vivo. This study provides a potential method of preparing a bioinspired stratified scaffold for the treatment of growth plate injuries.
Collapse
Affiliation(s)
- Xianggang Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - Zuhao Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - Jiaqi Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - Chenyu Wang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, 130021, PR China
| | - Haotian Bai
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - Xiujie Zhu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - Hui Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - Zhonghan Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - He Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - Jincheng Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| |
Collapse
|
30
|
Yang Y, Qiu B, Zhou Z, Hu C, Li J, Zhou C. Three-Dimensional Printing of Polycaprolactone/Nano-Hydroxyapatite Composite Scaffolds with a Pore Size of 300/500 µm is Histocompatible and Promotes Osteogenesis Using Rabbit Cortical Bone Marrow Stem Cells. Ann Transplant 2023; 28:e940365. [PMID: 37904328 PMCID: PMC10625337 DOI: 10.12659/aot.940365] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/12/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Many patients have bone defects that exceed the healing size. This study aimed to construct polycaprolactone/nano-hydroxyapatite (PCL/nHA) composite scaffolds with different pore sizes and investigate the osteogenesis and histocompatibility of cortical bone mesenchymal stem cells (BMSCs-C) seeded on it after inoculation. MATERIAL AND METHODS After mixing PCL and nHA proportionally, three-dimensional (3D) printing was used to print scaffolds. Porosity, compressive strength, and elastic modulus of PCL/nHA scaffolds were tested. The proliferation of BMSCs-C cells was examined and osteogenesis, chondrogenesis, and adipogenesis were evaluated. BMSCs-C cells were inoculated into 3D printing scaffolds, and histocompatibility between BMSCs-C cells and scaffolds was observed by the cell count kit (CCK-8) assay and LIVE/DEAD staining. After inoculating BMSCs-C cells into scaffolds, alkaline phosphatase (ALP) activity and calcium content were measured. RESULTS There was no obvious difference in characteristics between the 3 PCL/nHA composite scaffolds. The porosity, compressive strength, and elastic modulus of the 300/500-μm scaffold were between those of the 300-μm and 500-μm scaffolds. With increasing pore size, the mechanical properties of the scaffold decrease. BMSCs-C cells demonstrated faster growth and better osteogenic, adipogenic, and chondrogenic differentiation; therefore, BMSCs-C cells were selected as seed cells. PCL/nHA composite scaffolds with different pore sizes had no obvious toxicity and demonstrated good biocompatibility. All scaffolds showed higher ALP activity and calcium content. CONCLUSIONS The 300/500 μm mixed pore size scaffold took into account the mechanical properties of the 300 μm scaffold and the cell culture area of the 500 μm scaffold, therefore, 300/500 μm scaffold is a better model for the construction of tissue engineering scaffolds.
Collapse
|
31
|
Pan Q, Su W, Yao Y. Progress in microsphere-based scaffolds in bone/cartilage tissue engineering. Biomed Mater 2023; 18:062004. [PMID: 37751762 DOI: 10.1088/1748-605x/acfd78] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/26/2023] [Indexed: 09/28/2023]
Abstract
Bone/cartilage repair and regeneration have been popular and difficult issues in medical research. Tissue engineering is rapidly evolving to provide new solutions to this problem, and the key point is to design the appropriate scaffold biomaterial. In recent years, microsphere-based scaffolds have been considered suitable scaffold materials for bone/cartilage injury repair because microporous structures can form more internal space for better cell proliferation and other cellular activities, and these composite scaffolds can provide physical/chemical signals for neotissue formation with higher efficiency. This paper reviews the research progress of microsphere-based scaffolds in bone/chondral tissue engineering, briefly introduces types of microspheres made from polymer, inorganic and composite materials, discusses the preparation methods of microspheres and the exploration of suitable microsphere pore size in bone and cartilage tissue engineering, and finally details the application of microsphere-based scaffolds in biomimetic scaffolds, cell proliferation and drug delivery systems.
Collapse
Affiliation(s)
- Qian Pan
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Weixian Su
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Yongchang Yao
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| |
Collapse
|
32
|
Schynkel L, Meeremans M, Meyer AA, Schoolaert E, Geltmeyer J, Omidinia-Anarkoli A, Van Vlierberghe S, Daelemans L, De Laporte L, De Schauwer C, Hoogenboom R, De Clerck K. Cell Guiding Multicomponent Nanoyarn Tendon Scaffolds with Tunable Morphology and Flexibility. ACS APPLIED MATERIALS & INTERFACES 2023; 15:42241-42250. [PMID: 37650520 DOI: 10.1021/acsami.3c08241] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Nanofibrous scaffolds are widely investigated for tendon tissue engineering due to their porous structure, high flexibility, and the ability to guide cells in a preferred direction. Previous research has shown that providing a microenvironment similar to in vivo settings improves tissue regeneration. Therefore, in this work, ingenious multicomponent nanoyarn scaffolds that mimic the fibrillar and tubular structures of tendons are developed for the first time through electrospinning and bundling nanoyarns followed by electrospinning of a nanofibrous shell around the bundle. Multicomponent nanoyarn scaffolds out of poly(ε-caprolactone) with varying porosity, density, and diameter were successfully produced by coelectrospinning with water-soluble poly(2-ethyl-2-oxazoline) as a sacrificial component. The diameter and fiber orientation of the nanoyarns were successfully tuned based on parameter-morphology models obtained by the design of experiments. Cyclic bending tests were performed, indicating that the flexibility of the multicomponent nanoyarn scaffolds depends on the morphology and can be tuned through controlling the number of nanoyarns in the bundle and the porosity. Indirect and direct cell culture tests using mouse and equine tendon cells revealed excellent cytocompatibility of the nanofibrous products and demonstrated the potential of the nanoyarns to guide the growing cells along the nanofiber direction, which is crucial for tendon tissue engineering.
Collapse
Affiliation(s)
- Lucas Schynkel
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| | - Marguerite Meeremans
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Anna A Meyer
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstrasse. 50, 52074 Aachen, Germany
- Institute of Technical and Macromolecular Chemistry (ITMC), RWTH University Aachen, Worringerweg 2, 52074 Aachen ,Germany
| | - Ella Schoolaert
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| | - Jozefien Geltmeyer
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| | | | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281 - Building S4, 9000 Ghent, Belgium
| | - Lode Daelemans
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| | - Laura De Laporte
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstrasse. 50, 52074 Aachen, Germany
- Institute of Technical and Macromolecular Chemistry (ITMC), RWTH University Aachen, Worringerweg 2, 52074 Aachen ,Germany
- Advanced Materials for Biomedicine (AMB), Institute of Applied Medical Engineering (AME), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Forckenbeckstrasse 55, 52074 Aachen ,Germany
| | - Catharina De Schauwer
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281 - Building S4, 9000 Ghent, Belgium
| | - Karen De Clerck
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| |
Collapse
|
33
|
Blaudez F, Ivanovski S, Fernandez T, Vaquette C. Effect of In Vitro Culture Length on the Bone-Forming Capacity of Osteoblast-Derived Decellularized Extracellular Matrix Melt Electrowritten Scaffolds. Biomacromolecules 2023; 24:3450-3462. [PMID: 37458386 DOI: 10.1021/acs.biomac.2c01504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Recent advancements in decellularization have seen the development of extracellular matrix (ECM)-decorated scaffolds for bone regeneration; however, little is understood of the impact of in vitro culture prior to decellularization on the performances of these constructs. Therefore, this study investigated the effect of in vitro culture on ECM-decorated melt electrowritten polycaprolactone scaffold bioactivity. The scaffolds were seeded with osteoblasts and cultured for 1, 2, or 4 weeks to facilitate bone-specific ECM deposition and subsequently decellularized to form an acellular ECM-decorated scaffold. The utilization of mild chemicals and DNase was highly efficient in removing DNA while preserving ECM structure and composition. ECM decoration of the melt electrowritten fibers was observed within the first week of culture, with increased ECM at 2 and 4 week culture periods. Infiltration of re-seeded cells as well as overall bone regeneration in a rodent calvarial model was impeded by a longer culture period. Thus, it was demonstrated that the length of culture has a key influence on the osteogenic properties of decellularized ECM-decorated scaffolds, with long-term culture (2+ weeks) causing pore obstruction and creating a physical barrier which interfered with bone formation. These findings have important implications for the development of effective ECM-decorated scaffolds for bone regeneration.
Collapse
Affiliation(s)
- Fanny Blaudez
- School of Dentistry and Oral Health, Griffith University, Parklands Dr., Southport QLD 4222, Australia
- The University of Queensland, School of Dentistry, 288 Herston Rd., Herston QLD 4006, Australia
| | - Saso Ivanovski
- The University of Queensland, School of Dentistry, 288 Herston Rd., Herston QLD 4006, Australia
| | - Tulio Fernandez
- The University of Queensland, School of Dentistry, 288 Herston Rd., Herston QLD 4006, Australia
- College of Medicine and Dentistry, James Cook University, Cairns Campus, Cairns 4870, Australia
| | - Cedryck Vaquette
- The University of Queensland, School of Dentistry, 288 Herston Rd., Herston QLD 4006, Australia
| |
Collapse
|
34
|
Loi G, Scocozza F, Aliberti F, Rinvenuto L, Cidonio G, Marchesi N, Benedetti L, Ceccarelli G, Conti M. 3D Co-Printing and Substrate Geometry Influence the Differentiation of C2C12 Skeletal Myoblasts. Gels 2023; 9:595. [PMID: 37504474 PMCID: PMC10378771 DOI: 10.3390/gels9070595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/07/2023] [Accepted: 07/22/2023] [Indexed: 07/29/2023] Open
Abstract
Cells are influenced by several biomechanical aspects of their microenvironment, such as substrate geometry. According to the literature, substrate geometry influences the behavior of muscle cells; in particular, the curvature feature improves cell proliferation. However, the effect of substrate geometry on the myogenic differentiation process is not clear and needs to be further investigated. Here, we show that the 3D co-printing technique allows the realization of substrates. To test the influence of the co-printing technique on cellular behavior, we realized linear polycaprolactone substrates with channels in which a fibrinogen-based hydrogel loaded with C2C12 cells was deposited. Cell viability and differentiation were investigated up to 21 days in culture. The results suggest that this technology significantly improves the differentiation at 14 days. Therefore, we investigate the substrate geometry influence by comparing three different co-printed geometries-linear, circular, and hybrid structures (linear and circular features combined). Based on our results, all structures exhibit optimal cell viability (>94%), but the linear pattern allows to increase the in vitro cell differentiation, in particular after 14 days of culture. This study proposes an endorsed approach for creating artificial muscles for future skeletal muscle tissue engineering applications.
Collapse
Affiliation(s)
- Giada Loi
- Department of Civil Engineering and Architecture, University of Pavia, Via Adolfo Ferrata 3, 27100 Pavia, Italy
| | - Franca Scocozza
- Department of Civil Engineering and Architecture, University of Pavia, Via Adolfo Ferrata 3, 27100 Pavia, Italy
| | - Flaminia Aliberti
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
- Fondazione IRCCS Policlinico San Matteo, Center for Inherited Cardiovascular Diseases, Transplant Research Area, 27100 Pavia, Italy
| | - Lorenza Rinvenuto
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Gianluca Cidonio
- Center for Life Nano- & Neuro-Science (CLN2S), Fondazione Istituto Italiano di Tecnologia, 00161 Rome, Italy
| | - Nicola Marchesi
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Laura Benedetti
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Gabriele Ceccarelli
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, Via Adolfo Ferrata 3, 27100 Pavia, Italy
| |
Collapse
|
35
|
Yu L, Cavelier S, Hannon B, Wei M. Recent development in multizonal scaffolds for osteochondral regeneration. Bioact Mater 2023; 25:122-159. [PMID: 36817819 PMCID: PMC9931622 DOI: 10.1016/j.bioactmat.2023.01.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/30/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Osteochondral (OC) repair is an extremely challenging topic due to the complex biphasic structure and poor intrinsic regenerative capability of natural osteochondral tissue. In contrast to the current surgical approaches which yield only short-term relief of symptoms, tissue engineering strategy has been shown more promising outcomes in treating OC defects since its emergence in the 1990s. In particular, the use of multizonal scaffolds (MZSs) that mimic the gradient transitions, from cartilage surface to the subchondral bone with either continuous or discontinuous compositions, structures, and properties of natural OC tissue, has been gaining momentum in recent years. Scrutinizing the latest developments in the field, this review offers a comprehensive summary of recent advances, current hurdles, and future perspectives of OC repair, particularly the use of MZSs including bilayered, trilayered, multilayered, and gradient scaffolds, by bringing together onerous demands of architecture designs, material selections, manufacturing techniques as well as the choices of growth factors and cells, each of which possesses its unique challenges and opportunities.
Collapse
Affiliation(s)
- Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Sacha Cavelier
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Brett Hannon
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
| | - Mei Wei
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
- Department of Mechanical Engineering, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
36
|
Ye YJ, Xu YF, Hou YB, Yin DC, Su DB, Zhao ZX. The regulation of tendon stem cell distribution, morphology, and gene expression by the modulus of microfibers. Colloids Surf B Biointerfaces 2023; 228:113393. [PMID: 37327653 DOI: 10.1016/j.colsurfb.2023.113393] [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: 11/24/2022] [Revised: 05/21/2023] [Accepted: 06/04/2023] [Indexed: 06/18/2023]
Abstract
The mechanical properties of a stem cell culture substrate significantly impact cell adhesion, survival, migration, proliferation, and differentiation in vitro. A major challenge in engineering artificial stem cell substrate is to properly identify the relevant physical features of native stem cell niches, which are likely different for each stem cell type. The behavior of tendon stem cells has potentially significant implications for tendon repair. Here, microfiber scaffolds with various modulus of elasticity are fabricated by near-field electrospinning, and their regulating effects on the in vitro behavior of tendon stem cells (TSCs) are discussed in this study. The number of pseudopodia shows a biphasic relationship with the modulus of scaffold. The proliferation, polarization ratio and alignment degree along the fibers of the TSCs increase with the increase of fiber modulus. TSCs cultured on the scaffold with moderate modulus (1429 MPa) show the upregulation of tendon-specific genes (Col-I, Tnmd, SCX and TNCF). These microfiber scaffolds provide great opportunities to modulate TSCs behavior at the micrometer scales. In conclusion, this study provides an instructive mechanical microenvironment for TSCs behaviors and may lead to the development of desirable engineered artificial stem cell substrate for tendon healing.
Collapse
Affiliation(s)
- Ya-Jing Ye
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, ShaanXi 710072, PR China.
| | - Yi-Fan Xu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, ShaanXi 710072, PR China
| | - Ya-Bo Hou
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, ShaanXi 710072, PR China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, ShaanXi 710072, PR China.
| | - Dan-Bo Su
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, ShaanXi 710072, PR China
| | - Zi-Xu Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, ShaanXi 710072, PR China
| |
Collapse
|
37
|
Ye R, Liu S, Zhu W, Li Y, Huang L, Zhang G, Zhang Y. Synthesis, Characterization, Properties, and Biomedical Application of Chitosan-Based Hydrogels. Polymers (Basel) 2023; 15:2482. [PMID: 37299281 PMCID: PMC10255636 DOI: 10.3390/polym15112482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The prospective applications of chitosan-based hydrogels (CBHs), a category of biocompatible and biodegradable materials, in biomedical disciplines such as tissue engineering, wound healing, drug delivery, and biosensing have garnered great interest. The synthesis and characterization processes used to create CBHs play a significant role in determining their characteristics and effectiveness. The qualities of CBHs might be greatly influenced by tailoring the manufacturing method to get certain traits, including porosity, swelling, mechanical strength, and bioactivity. Additionally, characterization methods aid in gaining access to the microstructures and properties of CBHs. Herein, this review provides a comprehensive assessment of the state-of-the-art with a focus on the affiliation between particular properties and domains in biomedicine. Moreover, this review highlights the beneficial properties and wide application of stimuli-responsive CBHs. The main obstacles and prospects for the future of CBH development for biomedical applications are also covered in this review.
Collapse
Affiliation(s)
- Ruixi Ye
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.Y.); (S.L.); (W.Z.); (Y.L.); (G.Z.)
| | - Siyu Liu
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.Y.); (S.L.); (W.Z.); (Y.L.); (G.Z.)
| | - Wenkai Zhu
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.Y.); (S.L.); (W.Z.); (Y.L.); (G.Z.)
| | - Yurong Li
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.Y.); (S.L.); (W.Z.); (Y.L.); (G.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Long Huang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, 299 Bayi Road, Wuhan 430072, China;
| | - Guozheng Zhang
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.Y.); (S.L.); (W.Z.); (Y.L.); (G.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yeshun Zhang
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.Y.); (S.L.); (W.Z.); (Y.L.); (G.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
- Zhenjiang Zhongnong Biotechnology Co., Ltd., Zhenjiang 212121, China
| |
Collapse
|
38
|
Wong PY, Soo S, Wong ESC, Praveen P, Clode P, Baker MV, Matsubara VH. A Novel Antimicrobial Hydrogel for the Management of Periodontal Diseases. Int Dent J 2023; 73:354-361. [PMID: 36754776 DOI: 10.1016/j.identj.2023.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 02/09/2023] Open
Abstract
OBJECTIVES This study aimed to synthesise a drug-delivery system based on a porous polymer hydrogel, with antimicrobial properties against Porphyromonas gingivalis and potential to be used in tissue regeneration. MATERIAL AND METHODS 2-Hydroxyethyl methacrylate monomers were polymerised using thermal and photoactivation in the presence of silver nitrate (AgNO3) and/or chlorhexidine digluconate. Poly-2-hydroxyethyl methacrylate (pHEMA) hydrogels containing silver nanoparticles (AgNPs) and/or 0.12% chlorhexidine (CHX) were produced and characterised using cryo-SEM and confocal microscopy. Hydrogel degradation and leaching of AgNP were tested for 1.5 months. The antimicrobial properties were tested against P. gingivalis using broth culture system and disk diffusion tests. RESULTS Our methodology manufactured porous polymeric hydrogels doped with AgNPs and CHX. Hydrogels showed a successful delivery of CHX and sustainable release of AgNPs in a steady hydrogel degradation rate determined based on the weight loss of samples. Hydrogels with AgNPs or CHX had a significant antimicrobial effect against P. gingivalis, with CHX-hydrogels exhibiting a stronger effect than AgNP-hydrogels in the short-term assessment. AgNP-CHX hydrogels showed a compounded antimicrobial effect, whereas control hydrogels containing neither AgNPs nor CHX had no influence on bacterial growth (P < .05). CONCLUSIONS The dual-cured pHEMA hydrogel loaded with antimicrobial agents proved to be an efficient drug-delivery system against periodontopathogens, with the potential to be used as a scaffold for tissue regeneration.
Collapse
Affiliation(s)
- Pauline Yang Wong
- UWA Dental School, The University of Western Australia, Perth, Western Australia, Australia
| | - Shane Soo
- UWA Dental School, The University of Western Australia, Perth, Western Australia, Australia
| | - Edmund Soon-Chern Wong
- UWA Dental School, The University of Western Australia, Perth, Western Australia, Australia
| | - Praveen Praveen
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Peta Clode
- Centre for Microscopy, Characterisation, and Analysis and School of Biological Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Murray V Baker
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Victor Haruo Matsubara
- UWA Dental School, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
39
|
Zdraveva E, Bendelja K, Bočkor L, Dolenec T, Mijović B. Detection of Limbal Stem Cells Adhered to Melt Electrospun Silk Fibroin and Gelatin-Modified Polylactic Acid Scaffolds. Polymers (Basel) 2023; 15:polym15030777. [PMID: 36772078 PMCID: PMC9919663 DOI: 10.3390/polym15030777] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Limbal stem cells (LSCs) are of paramount importance in corneal epithelial tissue repair. The cornea becomes opaque in case of limbal stem cell deficiency (LSCD), which may cause serious damage to the ocular visual function. There are many techniques to restore damaged epithelium, one of which is the transplantation of healthy cultured LSCs, usually onto a human amniotic membrane or onto bio-based engineered scaffolds in recent years. In this study, melt electrospun polylactic acid (PLA) was modified by silk fibroin or gelatin and further cultured with LSCs originating from three different donors. In terms of physicochemical properties, both modifications slightly increased PLA scaffold porosity (with a significantly larger pore area for the PLA/gelatin) and improved the scaffolds' swelling percentage, as well as their biodegradation rate. In terms of the scaffold application function, the aim was to detect/visualize whether LSCs adhered to the scaffolds and to further determine cell viability (total number), as well as to observe p63 and CK3 expressions in the LSCs. LSCs were attached to the surface of microfibers, showing flattened conformations or 3D spheres in the formation of colonies or agglomerations, respectively. All scaffolds showed the ability to bind the cells onto the surface of individual microfibers (PLA and PLA/gelatin), or in between the microfibers (PLA/silk fibroin), with the latter showing the most intense red fluorescence of the stained cells. All scaffolds proved to be biocompatible, while the PLA/silk fibroin scaffolds showed the highest 98% viability of 2.9 × 106 LSCs, with more than 98% of p63 and less than 20% of CK3 expressions in the LSCs, thus confirming the support of their growth, proliferation and corneal epithelial differentiation. The results show the potential of these bio-engineered scaffolds to be used as an alternative clinical approach.
Collapse
Affiliation(s)
- Emilija Zdraveva
- Department of Fundamental Natural and Engineering Sciences, Faculty of Textile Technology, University of Zagreb, 10000 Zagreb, Croatia
| | - Krešo Bendelja
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Luka Bočkor
- Center for Applied Bioanthropology, Institute for Anthropological Research, 10000 Zagreb, Croatia
| | - Tamara Dolenec
- Department of Transfusion and Regenerative Medicine, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
| | - Budimir Mijović
- Department of Fundamental Natural and Engineering Sciences, Faculty of Textile Technology, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence:
| |
Collapse
|
40
|
Chauhan A, Bhatt AD. A review on design of scaffold for osteoinduction: Toward the unification of independent design variables. Biomech Model Mechanobiol 2023; 22:1-21. [PMID: 36121530 DOI: 10.1007/s10237-022-01635-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022]
Abstract
Biophysical stimulus quantifies the osteoinductivity of the scaffold concerning the mechanoregulatory mathematical models of scaffold-assisted cellular differentiation. Consider a set of independent structural variables ($) that comprises bulk porosity levels ([Formula: see text]) and a set of morphological features of the micro-structure ([Formula: see text]) associated with scaffolds, i.e., [Formula: see text]. The literature suggests that biophysical stimulus ([Formula: see text]) is a function of independent structural variables ($). Limited understanding of the functional correlation between biophysical stimulus and structural features results in the lack of the desired osteoinductivity in a scaffold. Consequently, it limits their broad applicability to assist bone tissue regeneration for treating critical-sized bone fractures. The literature indicates the existence of multi-dimensional independent design variable space as a probable reason for the general lack of osteoinductivity in scaffolds. For instance, known morphological features are the size, shape, orientation, continuity, and connectivity of the porous regions in the scaffold. It implies that the number of independent variables ([Formula: see text]) is more than two, i.e., [Formula: see text], which interact and influence the magnitude of [Formula: see text] in a unified manner. The efficiency of standard engineering design procedures to analyze the correlation between dependent variable ([Formula: see text]) and independent variables ($) in 3D mutually orthogonal Cartesian coordinate system diminishes proportionally with the increase in the number of independent variables ([Formula: see text]) (Deb in Optimization for engineering design-algorithms and examples, PHI Learning Private Limited, New Delhi, 2012). Therefore, there is an immediate need to devise a framework that has the potential to quantify the micro-structural's morphological features in a unified manner to increase the prospects of scaffold-assisted bone tissue regeneration.
Collapse
Affiliation(s)
- Atul Chauhan
- Department of Mechanical Engineering, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India.
| | - Amba D Bhatt
- Department of Mechanical Engineering, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India
| |
Collapse
|
41
|
Tolabi H, Davari N, Khajehmohammadi M, Malektaj H, Nazemi K, Vahedi S, Ghalandari B, Reis RL, Ghorbani F, Oliveira JM. Progress of Microfluidic Hydrogel-Based Scaffolds and Organ-on-Chips for the Cartilage Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2208852. [PMID: 36633376 DOI: 10.1002/adma.202208852] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/09/2022] [Indexed: 05/09/2023]
Abstract
Cartilage degeneration is among the fundamental reasons behind disability and pain across the globe. Numerous approaches have been employed to treat cartilage diseases. Nevertheless, none have shown acceptable outcomes in the long run. In this regard, the convergence of tissue engineering and microfabrication principles can allow developing more advanced microfluidic technologies, thus offering attractive alternatives to current treatments and traditional constructs used in tissue engineering applications. Herein, the current developments involving microfluidic hydrogel-based scaffolds, promising structures for cartilage regeneration, ranging from hydrogels with microfluidic channels to hydrogels prepared by the microfluidic devices, that enable therapeutic delivery of cells, drugs, and growth factors, as well as cartilage-related organ-on-chips are reviewed. Thereafter, cartilage anatomy and types of damages, and present treatment options are briefly overviewed. Various hydrogels are introduced, and the advantages of microfluidic hydrogel-based scaffolds over traditional hydrogels are thoroughly discussed. Furthermore, available technologies for fabricating microfluidic hydrogel-based scaffolds and microfluidic chips are presented. The preclinical and clinical applications of microfluidic hydrogel-based scaffolds in cartilage regeneration and the development of cartilage-related microfluidic chips over time are further explained. The current developments, recent key challenges, and attractive prospects that should be considered so as to develop microfluidic systems in cartilage repair are highlighted.
Collapse
Affiliation(s)
- Hamidreza Tolabi
- New Technologies Research Center (NTRC), Amirkabir University of Technology, Tehran, 15875-4413, Iran
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, 15875-4413, Iran
| | - Niyousha Davari
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran
| | - Mehran Khajehmohammadi
- Department of Mechanical Engineering, Faculty of Engineering, Yazd University, Yazd, 89195-741, Iran
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, 8916877391, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg, 9220, Denmark
| | - Katayoun Nazemi
- Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Samaneh Vahedi
- Department of Material Science and Engineering, Faculty of Engineering, Imam Khomeini International University, Qazvin, 34149-16818, Iran
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4805-017, Portugal
| | - Farnaz Ghorbani
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058, Erlangen, Germany
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4805-017, Portugal
| |
Collapse
|
42
|
Dozzo A, Chullipalliyalil K, McAuliffe M, O’Driscoll CM, Ryan KB. Nano-Hydroxyapatite/PLGA Mixed Scaffolds as a Tool for Drug Development and to Study Metastatic Prostate Cancer in the Bone. Pharmaceutics 2023; 15:pharmaceutics15010242. [PMID: 36678871 PMCID: PMC9864166 DOI: 10.3390/pharmaceutics15010242] [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: 12/08/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
(1) Background: Three-dimensional (3D) in vitro, biorelevant culture models that recapitulate cancer progression can help elucidate physio-pathological disease cues and enhance the screening of more effective therapies. Insufficient research has been conducted to generate in vitro 3D models to replicate the spread of prostate cancer to the bone, a key metastatic site of the disease, and to understand the interplay between the key cell players. In this study, we aim to investigate PLGA and nano-hydroxyapatite (nHA)/PLGA mixed scaffolds as a predictive preclinical tool to study metastatic prostate cancer (mPC) in the bone and reduce the gap that exists with traditional 2D cultures. (2) Methods: nHA/PLGA mixed scaffolds were produced by electrospraying, compacting, and foaming PLGA polymer microparticles, +/- nano-hydroxyapatite (nHA), and a salt porogen to produce 3D, porous scaffolds. Physicochemical scaffold characterisation together with an evaluation of osteoblastic (hFOB 1.19) and mPC (PC-3) cell behaviour (RT-qPCR, viability, and differentiation) in mono- and co-culture, was undertaken. (3) Results: The results show that the addition of nHA, particularly at the higher-level impacted scaffolds in terms of mechanical and degradation behaviour. The nHA 4 mg resulted in weaker scaffolds, but cell viability increased. Qualitatively, fluorescent imaging of cultures showed an increase in PC-3 cells compared to osteoblasts despite lower initial PC-3 seeding densities. Osteoblast monocultures, in general, caused an upregulation (or at least equivalent to controls) in gene production, which was highest in plain scaffolds and decreased with increases in nHA. Additionally, the genes were downregulated in PC3 and co-cultures. Further, drug toxicity tests demonstrated a significant effect in 2D and 3D co-cultures. (4) Conclusions: The results demonstrate that culture conditions and environment (2D versus 3D, monoculture versus co-culture) and scaffold composition all impact cell behaviour and model development.
Collapse
Affiliation(s)
- Annachiara Dozzo
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, T12 K8AF Cork, Ireland
| | | | - Michael McAuliffe
- Centre for Advanced Photonics & Process Analysis, Munster Technological University Cork, T12 P928 Cork, Ireland
| | - Caitriona M. O’Driscoll
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, T12 K8AF Cork, Ireland
| | - Katie B. Ryan
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, T12 K8AF Cork, Ireland
- Correspondence:
| |
Collapse
|
43
|
Xin L, Wen Y, Song J, Chen T, Zhai Q. Bone regeneration strategies based on organelle homeostasis of mesenchymal stem cells. Front Endocrinol (Lausanne) 2023; 14:1151691. [PMID: 37033227 PMCID: PMC10081449 DOI: 10.3389/fendo.2023.1151691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
The organelle modulation has emerged as a crucial contributor to the organismal homeostasis. The mesenchymal stem cells (MSCs), with their putative functions in maintaining the regeneration ability of adult tissues, have been identified as a major driver to underlie skeletal health. Bone is a structural and endocrine organ, in which the organelle regulation on mesenchymal stem cells (MSCs) function has most been discovered recently. Furthermore, potential treatments to control bone regeneration are developing using organelle-targeted techniques based on manipulating MSCs osteogenesis. In this review, we summarize the most current understanding of organelle regulation on MSCs in bone homeostasis, and to outline mechanistic insights as well as organelle-targeted approaches for accelerated bone regeneration.
Collapse
Affiliation(s)
- Liangjing Xin
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yao Wen
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- *Correspondence: Qiming Zhai, ; Tao Chen, ; Jinlin Song,
| | - Tao Chen
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- *Correspondence: Qiming Zhai, ; Tao Chen, ; Jinlin Song,
| | - Qiming Zhai
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- *Correspondence: Qiming Zhai, ; Tao Chen, ; Jinlin Song,
| |
Collapse
|
44
|
Wang J, Gao L, Song J, Li S. Study of EGCG composite hydrogel for the treatment of radiation-induced skin injuries. J Appl Biomater Funct Mater 2023; 21:22808000231218996. [PMID: 38131321 DOI: 10.1177/22808000231218996] [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] [Indexed: 12/23/2023] Open
Abstract
With the rising incidence of cancer, radiotherapy has become an increasingly popular treatment modality. However, radiation-induced skin injuries (RSIs) have emerged as a commonly reported side effect of radiotherapy, thereby presenting a significant challenge in the field of radiotherapy. In this study, we report the successful synthesis of a photosensitive hydrogel via amide reaction for grafting the photosensitive group, methacrylate anhydride (MA), onto chitosan (CHI) and gelatin (GEL), with subsequent physical incorporation of epigallocatechin gallate (EGCG). The resulting composite photosensitive hydrogels exhibited favorable swelling properties, rheological properties, and biocompatibility, which promote angiogenesis and demonstrate notable therapeutic efficacy against RSIs. These findings provide valuable insights into the clinical utility of EGCG composite hydrogels for the effective management of RSIs.
Collapse
Affiliation(s)
- Jingying Wang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Lin Gao
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Jianbo Song
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
- Collaborative Innovation Center For Molecular Imaging, Shanxi Medical University, Taiyuan, People's Republic of China
| |
Collapse
|
45
|
Guler S, Eichholz K, Chariyev-Prinz F, Pitacco P, Aydin HM, Kelly DJ, Vargel İ. Biofabrication of Poly(glycerol sebacate) Scaffolds Functionalized with a Decellularized Bone Extracellular Matrix for Bone Tissue Engineering. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 10:bioengineering10010030. [PMID: 36671602 PMCID: PMC9854839 DOI: 10.3390/bioengineering10010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022]
Abstract
The microarchitecture of bone tissue engineering (BTE) scaffolds has been shown to have a direct effect on the osteogenesis of mesenchymal stem cells (MSCs) and bone tissue regeneration. Poly(glycerol sebacate) (PGS) is a promising polymer that can be tailored to have specific mechanical properties, as well as be used to create microenvironments that are relevant in the context of BTE applications. In this study, we utilized PGS elastomer for the fabrication of a biocompatible and bioactive scaffold for BTE, with tissue-specific cues and a suitable microstructure for the osteogenic lineage commitment of MSCs. In order to achieve this, the PGS was functionalized with a decellularized bone (deB) extracellular matrix (ECM) (14% and 28% by weight) to enhance its osteoinductive potential. Two different pore sizes were fabricated (small: 100-150 μm and large: 250-355 μm) to determine a preferred pore size for in vitro osteogenesis. The decellularized bone ECM functionalization of the PGS not only improved initial cell attachment and osteogenesis but also enhanced the mechanical strength of the scaffold by up to 165 kPa. Furthermore, the constructs were also successfully tailored with an enhanced degradation rate/pH change and wettability. The highest bone-inserted small-pore scaffold had a 12% endpoint weight loss, and the pH was measured at around 7.14. The in vitro osteogenic differentiation of the MSCs in the PGS-deB blends revealed a better lineage commitment of the small-pore-sized and 28% (w/w) bone-inserted scaffolds, as evidenced by calcium quantification, ALP expression, and alizarin red staining. This study demonstrates a suitable pore size and amount of decellularized bone ECM for osteoinduction via precisely tailored PGS elastomer BTE scaffolds.
Collapse
Affiliation(s)
- Selcan Guler
- Bioengineering Division, Institute of Science and Engineering, Hacettepe University, 06800 Ankara, Turkey
| | - Kian Eichholz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Farhad Chariyev-Prinz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Pierluca Pitacco
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Halil Murat Aydin
- Bioengineering Division, Institute of Science and Engineering, Hacettepe University, 06800 Ankara, Turkey
| | - Daniel J. Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 F6N2 Dublin, Ireland
- Department of Anatomy, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - İbrahim Vargel
- Bioengineering Division, Institute of Science and Engineering, Hacettepe University, 06800 Ankara, Turkey
- Department of Plastic and Reconstructive Surgery, Hacettepe University Hospitals, 06230 Ankara, Turkey
- Correspondence:
| |
Collapse
|
46
|
Passos M, Zankovic S, Minas G, Klüver E, Baltzer M, Schmal H, Seidenstuecker M. About 3D Printability of Thermoplastic Collagen for Biomedical Applications. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120780. [PMID: 36550986 PMCID: PMC9774095 DOI: 10.3390/bioengineering9120780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/22/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
With more than 1.5 million total knee and hip implants placed each year, there is an urgent need for a drug delivery system that can effectively support the repair of bone infections. Scaffolds made of natural biopolymers are widely used for this purpose due to their biocompatibility, biodegradability, and suitable mechanical properties. However, the poor processability is a bottleneck, as highly customizable scaffolds are desired. The aim of the present research is to develop a scaffold made of thermoplastic collagen (TC) using 3D printing technology. The viscosity of the material was measured using a rheometer. A 3D bioplotter was used to fabricate the scaffolds out of TC. The mechanical properties of the TC scaffolds were performed using tension/compression testing on a Zwick/Roell universal testing machine. TC shows better compressibility with increasing temperature and a decrease in dynamic viscosity (η), storage modulus (G'), and loss modulus (G″). The compressive strength of the TC scaffolds was between 3-10 MPa, depending on the geometry (cylinder or cuboid, with different infills). We have demonstrated for the first time that TC can be used to fabricate porous scaffolds by 3D printing in various geometries.
Collapse
Affiliation(s)
- Marina Passos
- G.E.R.N. Center of Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
- Center for Micro Electromechanical Systems (CMEMS-UMinho), University of Minho Campus de Azurém, 4800-058 Guimarães, Portugal
| | - Sergej Zankovic
- G.E.R.N. Center of Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Graça Minas
- LABBELS-Associate Laboratory, University of Minho Campus de Gualtar, 4710-057 Braga, Portugal
| | - Enno Klüver
- FILK Freiberg Institute gGmbH, Meissner Ring 1-5, 09599 Freiberg, Germany
| | - Marit Baltzer
- FILK Freiberg Institute gGmbH, Meissner Ring 1-5, 09599 Freiberg, Germany
| | - Hagen Schmal
- G.E.R.N. Center of Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Michael Seidenstuecker
- G.E.R.N. Center of Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
- Correspondence:
| |
Collapse
|
47
|
Shahverdi M, Seifi S, Akbari A, Mohammadi K, Shamloo A, Movahhedy MR. Melt electrowriting of PLA, PCL, and composite PLA/PCL scaffolds for tissue engineering application. Sci Rep 2022; 12:19935. [PMID: 36402790 PMCID: PMC9675866 DOI: 10.1038/s41598-022-24275-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022] Open
Abstract
Fabrication of well-ordered and bio-mimetic scaffolds is one of the most important research lines in tissue engineering. Different techniques have been utilized to achieve this goal, however, each method has its own disadvantages. Recently, melt electrowriting (MEW) as a technique for fabrication of well-organized scaffolds has attracted the researchers' attention due to simultaneous use of principles of additive manufacturing and electrohydrodynamic phenomena. In previous research studies, polycaprolactone (PCL) has been mostly used in MEW process. PCL is a biocompatible polymer with characteristics that make it easy to fabricate well-arranged structures using MEW device. However, the mechanical properties of PCL are not favorable for applications like bone tissue engineering. Furthermore, it is of vital importance to demonstrate the capability of MEW technique for processing a broad range of polymers. To address aforementioned problems, in this study, three ten-layered box-structured well-ordered scaffolds, including neat PLA, neat PCL, and PLA/PCL composite are fabricated using an MEW device. Printing of the composite PLA/PCL scaffold using the MEW device is conducted in this study for the first time. The MEW device used in this study is a commercial fused deposition modeling (FDM) 3D printer which with some changes in its setup and configuration becomes prepared for being used as an MEW device. Since in most of previous studies, a setup has been designed and built for MEW process, the use of the FDM device can be considered as one of the novelties of this research. The printing parameters are adjusted in a way that scaffolds with nearly equal pore sizes in the range of 140 µm to 150 µm are fabricated. However, PCL fibers are mostly narrower (diameters in the range of 5 µm to 15 µm) than PLA fibers with diameters between 15 and 25 µm. Unlike the MEW process of PCL, accurate positioning of PLA fibers is difficult which can be due to higher viscosity of PLA melt compared to PCL melt. The printed composite PLA/PCL scaffold possesses a well-ordered box structure with improved mechanical properties and cell-scaffold interactions compared to both neat PLA and PCL scaffolds. Besides, the composite scaffold exhibits a higher swelling ratio than the neat PCL scaffold which can be related to the presence of less hydrophobic PLA fibers. This scaffold demonstrates an anisotropic behavior during uniaxial tensile test in which its Young's modulus, ultimate tensile stress, and strain to failure all depend on the direction of the applied tensile force. This anisotropy makes the composite PLA/PCL scaffold an exciting candidate for applications in heart tissue engineering. The results of in-vitro cell viability test using L929 mouse murine fibroblast and human umbilical vein endothelial (HUVEC) cells demonstrate that all of the printed scaffolds are biocompatible. In particular, the composite scaffold presents the highest cell viability value among the fabricated scaffolds. All in all, the composite PLA/PCL scaffold shows that it can be a promising substitution for neat PCL scaffold used in previous MEW studies.
Collapse
Affiliation(s)
- Mohammad Shahverdi
- Advanced Manufacturing Laboratory, School of Mechanical Engineering, Sharif University of Technology, Azadi Ave., Tehran, Iran
| | - Saeed Seifi
- Nano BioTechnology Laboratory, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Ali Akbari
- Advanced Manufacturing Laboratory, School of Mechanical Engineering, Sharif University of Technology, Azadi Ave., Tehran, Iran
| | - Kaivan Mohammadi
- Advanced Manufacturing Laboratory, School of Mechanical Engineering, Sharif University of Technology, Azadi Ave., Tehran, Iran.
| | - Amir Shamloo
- Nano BioTechnology Laboratory, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran.
| | - Mohammad Reza Movahhedy
- Advanced Manufacturing Laboratory, School of Mechanical Engineering, Sharif University of Technology, Azadi Ave., Tehran, Iran
| |
Collapse
|
48
|
Kowalczyk P, Wojasiński M, Jaroszewicz J, Kopeć K, Ciach T. Controlled formation of highly porous polylactic acid‑calcium phosphate granules with defined structure. BIOMATERIALS ADVANCES 2022; 144:213195. [PMID: 36434927 DOI: 10.1016/j.bioadv.2022.213195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Synthetic bone repair materials are becoming increasingly popular in tissue engineering as a replacement for autografts and human/animal-based bone grafts. The biomedical application requires precise control over the material composition and structure, as well as over the size of granulate used for filling the bone defects, as the pore size and interconnectivity affect the regeneration process. This paper proposes a process of alloplastic and biodegradable polylactic acid/β-tricalcium phosphate granulates preparation and its parameters described. Using solvent-induced phase separation technique, porous spheres have been obtained in various sizes and morphologies. The design of the experiment's approach generated an experimental plan for further statistical modeling using the resulting data. The statistical modeling approach to the data from conducting a designed set of experiments allowed analysis of the influence of process parameters on the properties of the resulting granules. We confirmed that the content of β-tricalcium phosphate plays the most significant role in the size distribution of prepared granulate. The shape of the particles becomes less spherical with higher phosphate concentration in the emulsion. The proposed technique allows preparing porous granulates in the 0.2-1.8 mm diameter range, where granules' mean diameter and sphericity are tunable with polymer and phosphate concentrations. The granulate created a potentially implantable scaffold for resected bone regeneration, as cytotoxicity tests assured the material is non-cytotoxic in vitro, and human mesenchymal stem cells have been cultured on the surface of granulates. Results from cell cultures seeded on the Resomer LR 706S granulates were the most promising.
Collapse
Affiliation(s)
- Piotr Kowalczyk
- Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645 Warsaw, Poland.
| | - Michał Wojasiński
- Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645 Warsaw, Poland
| | - Jakub Jaroszewicz
- Faculty of Material Science and Engineering, Warsaw University of Technology, Wołoska 141, 02-507 Warsaw, Poland
| | - Kamil Kopeć
- Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645 Warsaw, Poland
| | - Tomasz Ciach
- Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645 Warsaw, Poland
| |
Collapse
|
49
|
Ma Y, Wang X, Su T, Lu F, Chang Q, Gao J. Recent Advances in Macroporous Hydrogels for Cell Behavior and Tissue Engineering. Gels 2022; 8:606. [PMID: 36286107 PMCID: PMC9601978 DOI: 10.3390/gels8100606] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
Hydrogels have been extensively used as scaffolds in tissue engineering for cell adhesion, proliferation, migration, and differentiation because of their high-water content and biocompatibility similarity to the extracellular matrix. However, submicron or nanosized pore networks within hydrogels severely limit cell survival and tissue regeneration. In recent years, the application of macroporous hydrogels in tissue engineering has received considerable attention. The macroporous structure not only facilitates nutrient transportation and metabolite discharge but also provides more space for cell behavior and tissue formation. Several strategies for creating and functionalizing macroporous hydrogels have been reported. This review began with an overview of the advantages and challenges of macroporous hydrogels in the regulation of cellular behavior. In addition, advanced methods for the preparation of macroporous hydrogels to modulate cellular behavior were discussed. Finally, future research in related fields was discussed.
Collapse
Affiliation(s)
| | | | | | | | - Qiang Chang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou 510515, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou 510515, China
| |
Collapse
|
50
|
Esmaeili J, Barati A, Charelli LE. Discussing the final size and shape of the reconstructed tissues in tissue engineering. J Artif Organs 2022:10.1007/s10047-022-01360-1. [PMID: 36125581 DOI: 10.1007/s10047-022-01360-1] [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: 04/05/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
Tissue engineering (TE) has made a revolution in repairing, replacing, or regenerating tissues or organs, but it has still a long way ahead. The mechanical properties along with suitable physicochemical and biological characteristics are the initial criteria for scaffolds in TE that should be fulfilled. This research will provide another point of view toward TE challenges concerning the morphological and geometrical aspects of the reconstructed tissue and which parameters may affect it. Based on our survey, there is a high possibility that the final reconstructed tissue may be different in size and shape compared to the original design scaffold. Thereby, the 3D-printed scaffold might not guarantee an accurate tissue reconstruction. The main justification for this is the unpredicted behavior of cells, specifically in the outer layer of the scaffold. It can also be a concern when the scaffold is implanted while cell migration cannot be controlled through the in vivo signaling pathways, which might cause cancer challenges. To sum up, it is concluded that more studies are necessary to focus on the size and geometry of the final reconstructed tissue.
Collapse
Affiliation(s)
- Javad Esmaeili
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, 38156-88349, Iran.,Tissue Engineering Department, TISSUEHUB Co., Tehran, Iran
| | - Aboulfazl Barati
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, 38156-88349, Iran.
| | - Letícia Emiliano Charelli
- Nanotechnology Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering, COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|