1
|
Piszko PJ, Piszko A, Kiryk S, Kiryk J, Horodniczy T, Struzik N, Wiśniewska K, Matys J, Dobrzyński M. Bone Regeneration Capabilities of Scaffolds Containing Chitosan and Nanometric Hydroxyapatite-Systematic Review Based on In Vivo Examinations. Biomimetics (Basel) 2024; 9:503. [PMID: 39194482 DOI: 10.3390/biomimetics9080503] [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: 07/10/2024] [Revised: 08/05/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
In this systematic review, the authors aimed to investigate the state of knowledge on in vivo evaluations of chitosan and nanometric hydroxyapatite (nanohydroxyapatite, nHAp) scaffolds for bone-tissue regeneration. In March 2024, an electronic search was systematically conducted across the PubMed, Cochrane, and Web of Science databases using the keywords (hydroxyapatite) AND (chitosan) AND (scaffold) AND (biomimetic). Methodologically, the systematic review followed the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) protocol to the letter. Initially, a total of 375 studies were screened, and 164 duplicates were removed. A further 188 articles were excluded because they did not correspond to the predefined topics, and an additional 3 articles were eliminated due to the inability to obtain the full text. The final compilation included 20 studies. All publications indicated a potential beneficial effect of the scaffolds in in vivo bone defect repair. A beneficial effect of hydroxyapatite as a scaffold component was observed in 16 studies, including greater mechanical resistance, cellular differentiation, and enhanced bone damage regeneration. The addition of chitosan and apatite ceramics, which combined the strengths of both materials, had the potential to become a useful bone-tissue engineering material.
Collapse
Affiliation(s)
- Paweł J Piszko
- Department of Pediatric Dentistry and Preclinical Dentistry, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland
| | - Aleksandra Piszko
- Department of Pediatric Dentistry and Preclinical Dentistry, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland
| | - Sylwia Kiryk
- Department of Pediatric Dentistry and Preclinical Dentistry, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland
| | - Jan Kiryk
- Department of Dental Surgery, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland
| | - Tomasz Horodniczy
- Ortho.pl Centrum Zdrowego Uśmiechu, Buforowa 34, 52-131 Wrocław, Poland
| | - Natalia Struzik
- Pre-Clinical Research Centre, Wroclaw Medical University, Bujwida 44, 50-368 Wrocław, Poland
| | - Kamila Wiśniewska
- Department of Dental Surgery, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland
| | - Jacek Matys
- Department of Dental Surgery, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland
| | - Maciej Dobrzyński
- Department of Pediatric Dentistry and Preclinical Dentistry, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland
| |
Collapse
|
2
|
Zhang Y, Chen J, Sun Y, Wang M, Liu H, Zhang W. Endogenous Tissue Engineering for Chondral and Osteochondral Regeneration: Strategies and Mechanisms. ACS Biomater Sci Eng 2024; 10:4716-4739. [PMID: 39091217 DOI: 10.1021/acsbiomaterials.4c00603] [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: 08/04/2024]
Abstract
Increasing attention has been paid to the development of effective strategies for articular cartilage (AC) and osteochondral (OC) regeneration due to their limited self-reparative capacities and the shortage of timely and appropriate clinical treatments. Traditional cell-dependent tissue engineering faces various challenges such as restricted cell sources, phenotypic alterations, and immune rejection. In contrast, endogenous tissue engineering represents a promising alternative, leveraging acellular biomaterials to guide endogenous cells to the injury site and stimulate their intrinsic regenerative potential. This review provides a comprehensive overview of recent advancements in endogenous tissue engineering strategies for AC and OC regeneration, with a focus on the tissue engineering triad comprising endogenous stem/progenitor cells (ESPCs), scaffolds, and biomolecules. Multiple types of ESPCs present within the AC and OC microenvironment, including bone marrow-derived mesenchymal stem cells (BMSCs), adipose-derived mesenchymal stem cells (AD-MSCs), synovial membrane-derived mesenchymal stem cells (SM-MSCs), and AC-derived stem/progenitor cells (CSPCs), exhibit the ability to migrate toward injury sites and demonstrate pro-regenerative properties. The fabrication and characteristics of scaffolds in various formats including hydrogels, porous sponges, electrospun fibers, particles, films, multilayer scaffolds, bioceramics, and bioglass, highlighting their suitability for AC and OC repair, are systemically summarized. Furthermore, the review emphasizes the pivotal role of biomolecules in facilitating ESPCs migration, adhesion, chondrogenesis, osteogenesis, as well as regulating inflammation, aging, and hypertrophy-critical processes for endogenous AC and OC regeneration. Insights into the applications of endogenous tissue engineering strategies for in vivo AC and OC regeneration are provided along with a discussion on future perspectives to enhance regenerative outcomes.
Collapse
Affiliation(s)
- Yanan Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| |
Collapse
|
3
|
Wei B, Xu Y, Tang C, Liu NQ, Li X, Yao Q, Wang L. An injectable active hydrogel based on BMSC-derived extracellular matrix for cartilage regeneration enhancement. BIOMATERIALS ADVANCES 2024; 160:213857. [PMID: 38657287 DOI: 10.1016/j.bioadv.2024.213857] [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: 01/02/2024] [Revised: 03/20/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024]
Abstract
Articular cartilage injury impairs joint function and necessitates orthopedic intervention to restore the structure and function of the cartilage. Extracellular matrix (ECM) scaffolds derived from bone marrow mesenchymal stem cells (BMSCs) can effectively promote cell adhesion, proliferation, and chondrogenesis. However, pre-shaped ECM scaffolds have limited applicability due to their poor fit with the irregular surface of most articular cartilage defects. In this study, we fabricated an injectable active ECM hydrogel from autologous BMSCs-derived ECM by freeze-drying, liquid nitrogen milling, and enzymatic digestion. Moreover, our in vitro and in vivo results demonstrated that the prepared hydrogel enhanced chondrocyte adhesion and proliferation, chondrogenesis, cartilage regeneration, and integration with host tissue, respectively. These findings indicate that active ECM components can provide trophic support for cell proliferation and differentiation, restoring the structure and function of damaged cartilage.
Collapse
Affiliation(s)
- Bo Wei
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| | - Yan Xu
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Cheng Tang
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Nancy Q Liu
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA 90007, USA
| | - Xuxiang Li
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Liming Wang
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| |
Collapse
|
4
|
Cheng C, Peng X, Luo Y, Shi S, Wang L, Wang Y, Yu X. A photocrosslinked methacrylated carboxymethyl chitosan/oxidized locust bean gum double network hydrogel for cartilage repair. J Mater Chem B 2023; 11:10464-10481. [PMID: 37901956 DOI: 10.1039/d3tb01701j] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Repairing articular cartilage defects is a great challenge due to the poor self-regenerative capability of cartilage. Inspired by active substances found in the natural cartilage extracellular matrix, we used methacrylated carboxymethyl chitosan (MA-CMCS) and oxidized locust bean gum (OLBG) as the hydrogel backbone, and prepared a photocrosslinked dual network hydrogel containing allicin and decellularized cartilage powder (DCP). The rheological, swelling and water retention capacities of MA-CMCS@OLBG-Allicin/DCP (MCOAC) hydrogels were investigated to confirm the successful preparation of hydrogels suitable for cartilage repair. The MCOAC hydrogels showed good antibacterial ability to kill S. aureus and E. coli and anti-inflammatory properties due to the introduction of allicin. Furthermore, MA-CMCS@OLBG-Allicin/DCP hydrogels presented good cytocompatibility due to the addition of DCP, which could promote chondrocyte proliferation and promote the differentiation of BMSCs to chondrocytes. Further studies in vivo demonstrated that the DCP-contained MCOAC hydrogel exhibited superior performance in promoting cartilage tissue growth and wound healing in articular cartilage defects. Thus, the MCOAC hydrogel is a promising cartilage repair hydrogel with potential for clinical use.
Collapse
Affiliation(s)
- Can Cheng
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Xu Peng
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
- Experimental and Research Animal Institute, Sichuan University, Chengdu 610065, P. R. China
| | - Yihao Luo
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Shubin Shi
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Ling Wang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Yuhang Wang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Xixun Yu
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| |
Collapse
|
5
|
Browe DC, Burdis R, Díaz-Payno PJ, Freeman FE, Nulty JM, Buckley CT, Brama PA, Kelly DJ. Promoting endogenous articular cartilage regeneration using extracellular matrix scaffolds. Mater Today Bio 2022; 16:100343. [PMID: 35865410 PMCID: PMC9294195 DOI: 10.1016/j.mtbio.2022.100343] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 12/13/2022] Open
Abstract
Articular cartilage defects fail to heal spontaneously, typically progressing to osteoarthritis. Bone marrow stimulation techniques such as microfracture (MFX) are the current surgical standard of care; however MFX typically produces an inferior fibro-cartilaginous tissue which provides only temporary symptomatic relief. Here we implanted solubilised articular cartilage extracellular matrix (ECM) derived scaffolds into critically sized chondral defects in goats, securely anchoring these implants to the joint surface using a 3D-printed fixation device that overcame the need for sutures or glues. In vitro these ECM scaffolds were found to be inherently chondro-inductive, while in vivo they promoted superior articular cartilage regeneration compared to microfracture. In an attempt to further improve the quality of repair, we loaded these scaffolds with a known chemotactic factor, transforming growth factor (TGF)-β3. In vivo such TGF-β3 loaded scaffolds promoted superior articular cartilage regeneration. This study demonstrates that ECM derived biomaterials, either alone and particularly when combined with exogenous growth factors, can successfully treat articular cartilage defects in a clinically relevant large animal model.
Collapse
Affiliation(s)
- David C. Browe
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Ireland
| | - Ross Burdis
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Pedro J. Díaz-Payno
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Fiona E. Freeman
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
- Institute for Medical Engineering and Science Massachusetts Institute of Technology Cambridge, MA, 02142, USA
- Department of Medicine Division of Engineering in Medicine Brigham and Women’s Hospital Harvard Medical School Boston, MA, 02115, USA
| | - Jessica M. Nulty
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Conor T. Buckley
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Pieter A.J. Brama
- Section of Veterinary Clinical Sciences, School of Veterinary Medicine, University College Dublin, Ireland
| | - Daniel J. Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
6
|
Levingstone TJ, Sheehy EJ, Moran CJ, Cunniffe GM, Diaz Payno PJ, Brady RT, Almeida HV, Carroll SF, O’Byrne JM, Kelly DJ, Brama PAJ, O’ Brien FJ. Evaluation of a co-culture of rapidly isolated chondrocytes and stem cells seeded on tri-layered collagen-based scaffolds in a caprine osteochondral defect model. BIOMATERIALS AND BIOSYSTEMS 2022; 8:100066. [PMID: 36824377 PMCID: PMC9934472 DOI: 10.1016/j.bbiosy.2022.100066] [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: 07/04/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 12/05/2022] Open
Abstract
Cartilage has poor regenerative capacity and thus damage to the joint surfaces presents a major clinical challenge. Recent research has focussed on the development of tissue-engineered and cell-based approaches for the treatment of cartilage and osteochondral injuries, with current clinically available cell-based approaches including autologous chondrocyte implantation and matrix-assisted autologous chondrocyte implantation. However, these approaches have significant disadvantages due to the requirement for a two-stage surgical procedure and an in vitro chondrocyte expansion phase which increases logistical challenges, hospital times and costs. In this study, we hypothesized that seeding biomimetic tri-layered scaffolds, with proven regenerative potential, with chondrocyte/infrapatellar fat pad stromal cell co-cultures would improve their regenerative capacity compared to scaffolds implanted cell-free. Rapid cell isolation techniques, without the requirement for long term in vitro culture, were utilised to achieve co-cultures of chondrocytes and stromal cells and thus overcome the limitations of existing cell-based techniques. Cell-free and cell-seeded scaffolds were implanted in osteochondral defects, created within the femoral condyle and trochlear ridge, in a translational large animal goat model. While analysis showed trends towards delayed subchondral bone healing in the cell-seeded scaffold group, by the 12 month timepoint the cell-free and cell-seeded groups yield cartilage and bone tissue with comparable quality and quantity. The results of the study reinforce the potential of the biomimetic tri-layered scaffold to repair joint defects but failed to demonstrate a clear benefit from the addition of the CC/FPMSC co-culture to this scaffold. Taking into consideration the additional cost and complexity associated with the cell-seeded scaffold approach, this study demonstrates that the treatment of osteochondral defects using cell-free tri-layered scaffolds may represent a more prudent clinical approach.
Collapse
Affiliation(s)
- Tanya J. Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland,Centre for Medical Engineering Research (MEDeng), Dublin City University, Dublin 9, Ireland,Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland,Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123St. Stephen's Green, Dublin 2, Ireland,Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland
| | - Eamon J. Sheehy
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123St. Stephen's Green, Dublin 2, Ireland,Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland
| | - Conor J. Moran
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123St. Stephen's Green, Dublin 2, Ireland,Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland
| | - Gráinne M. Cunniffe
- Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland,National Spinal Injuries Unit, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Pedro J. Diaz Payno
- Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Robert T. Brady
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123St. Stephen's Green, Dublin 2, Ireland,Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland
| | - Henrique V. Almeida
- Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland,iBET, Instituto de Biologia Experimental e Tecnológica, 2781-901 Oeiras, Portugal,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Simon F. Carroll
- Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - John M. O’Byrne
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123St. Stephen's Green, Dublin 2, Ireland,Cappagh National Orthopaedic Hospital, Finglas, Dublin 11, Ireland
| | - Daniel J. Kelly
- Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Pieter AJ. Brama
- Section Veterinary Clinical Sciences, School of Veterinary Medicine, University College Dublin, Dublin 4, Ireland
| | - Fergal J. O’ Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123St. Stephen's Green, Dublin 2, Ireland,Trinity Centre for Biomedical Engineering (TCBE), Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2, Ireland,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland,Corresponding author at: Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123St. Stephen's Green, Ireland
| |
Collapse
|
7
|
Jankauskaite L, Malinauskas M, Aukstikalne L, Dabasinskaite L, Rimkunas A, Mickevicius T, Pockevičius A, Krugly E, Martuzevicius D, Ciuzas D, Baniukaitiene O, Usas A. Functionalized Electrospun Scaffold-Human-Muscle-Derived Stem Cell Construct Promotes In Vivo Neocartilage Formation. Polymers (Basel) 2022; 14:polym14122498. [PMID: 35746068 PMCID: PMC9229929 DOI: 10.3390/polym14122498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 02/05/2023] Open
Abstract
Polycaprolactone (PCL) is a non-cytotoxic, completely biodegradable biomaterial, ideal for cartilage tissue engineering. Despite drawbacks such as low hydrophilicity and lack of functional groups necessary for incorporating growth factors, it provides a proper environment for different cells, including stem cells. In our study, we aimed to improve properties of scaffolds for better cell adherence and cartilage regeneration. Thus, electrospun PCL–scaffolds were functionalized with ozone and loaded with TGF-β3. Together, human-muscle-derived stem cells (hMDSCs) were isolated and assessed for their phenotype and potential to differentiate into specific lineages. Then, hMDSCs were seeded on ozonated (O) and non-ozonated (“naïve” (NO)) scaffolds with or without protein and submitted for in vitro and in vivo experiments. In vitro studies showed that hMDSC and control cells (human chondrocyte) could be tracked for at least 14 days. We observed better proliferation of hMDSCs in O scaffolds compared to NO scaffolds from day 7 to day 28. Protein analysis revealed slightly higher expression of type II collagen (Coll2) on O scaffolds compared to NO on days 21 and 28. We detected more pronounced formation of glycosaminoglycans in the O scaffolds containing TGF-β3 and hMDSC compared to NO and scaffolds without TGF-β3 in in vivo animal experiments. Coll2-positive extracellular matrix was observed within O and NO scaffolds containing TGF-β3 and hMDSC for up to 8 weeks after implantation. These findings suggest that ozone-treated, TGF-β3-loaded scaffold with hMDSC is a promising tool in neocartilage formation.
Collapse
Affiliation(s)
- Lina Jankauskaite
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
- Correspondence:
| | - Mantas Malinauskas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| | - Lauryna Aukstikalne
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| | - Lauryna Dabasinskaite
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Augustinas Rimkunas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| | - Tomas Mickevicius
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| | - Alius Pockevičius
- Pathology Center, Department of Veterinary Pathobiology, Veterinary Academy, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania;
| | - Edvinas Krugly
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Dainius Martuzevicius
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Darius Ciuzas
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Odeta Baniukaitiene
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Arvydas Usas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| |
Collapse
|
8
|
Browe DC, Díaz-Payno PJ, Freeman FE, Schipani R, Burdis R, Ahern DP, Nulty JM, Guler S, Randall LD, Buckley CT, Brama PA, Kelly DJ. Bilayered extracellular matrix derived scaffolds with anisotropic pore architecture guide tissue organization during osteochondral defect repair. Acta Biomater 2022; 143:266-281. [PMID: 35278686 DOI: 10.1016/j.actbio.2022.03.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/15/2022]
Abstract
While some clinical advances in cartilage repair have occurred, osteochondral (OC) defect repair remains a significant challenge, with current scaffold-based approaches failing to recapitulate the complex, hierarchical structure of native articular cartilage (AC). To address this need, we fabricated bilayered extracellular matrix (ECM)-derived scaffolds with aligned pore architectures. By modifying the freeze-drying kinetics and controlling the direction of heat transfer during freezing, it was possible to produce anisotropic scaffolds with larger pores which supported homogenous cellular infiltration and improved sulfated glycosaminoglycan deposition. Neo-tissue organization in vitro could also be controlled by altering scaffold pore architecture, with collagen fibres aligning parallel to the long-axis of the pores within scaffolds containing aligned pore networks. Furthermore, we used in vitro and in vivo assays to demonstrate that AC and bone ECM derived scaffolds could preferentially direct the differentiation of mesenchymal stromal cells (MSCs) towards either a chondrogenic or osteogenic lineage respectively, enabling the development of bilayered ECM scaffolds capable of spatially supporting unique tissue phenotypes. Finally, we implanted these scaffolds into a large animal model of OC defect repair. After 6 months in vivo, scaffold implantation was found to improve cartilage matrix deposition, with collagen fibres preferentially aligning parallel to the long axis of the scaffold pores, resulting in a repair tissue that structurally and compositionally was more hyaline-like in nature. These results demonstrate how scaffold architecture and composition can be spatially modulated to direct the regeneration of complex interfaces such as the osteochondral unit, enabling their use as cell-free, off-the-shelf implants for joint regeneration. STATEMENT OF SIGNIFICANCE: The architecture of the extracellular matrix, while integral to tissue function, is often neglected in the design and evaluation of regenerative biomaterials. In this study we developed a bilayered scaffold for osteochondral defect repair consisting of tissue-specific extracellular matrix (ECM)-derived biomaterials to spatially direct stem/progenitor cell differentiation, with a tailored pore microarchitecture to promote the development of a repair tissue that recapitulates the hierarchical structure of native AC. The use of this bilayered scaffold resulted in improved tissue repair outcomes in a large animal model, specifically the ability to guide neo-tissue organization and therefore recapitulate key aspects of the zonal structure of native articular cartilage. These bilayer scaffolds have the potential to become a new therapeutic option for osteochondral defect repair.
Collapse
|
9
|
Zare P, Pezeshki-Modaress M, Davachi SM, Chahsetareh H, Simorgh S, Asgari N, Haramshahi MA, Alizadeh R, Bagher Z, Farhadi M. An additive manufacturing-based 3D printed poly ɛ-caprolactone/alginate sulfate/extracellular matrix construct for nasal cartilage regeneration. J Biomed Mater Res A 2022; 110:1199-1209. [PMID: 35098649 DOI: 10.1002/jbm.a.37363] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/28/2021] [Accepted: 01/10/2022] [Indexed: 02/06/2023]
Abstract
Various composite scaffolds with different fabrication techniques have been applied in cartilage tissue engineering. In this study, poly ɛ-caprolactone (PCL) was printed by fused deposition modeling method, and the prepared scaffold was filled with Alginate (Alg): Alginate-Sulfate (Alg-Sul) hydrogel to provide a better biomimetic environment and emulate the structure of glycosaminoglycans properly. Furthermore, to enhance chondrogenesis, different concentrations of decellularized extracellular matrix (dECM) were added to the hydrogel. For cellular analyses, the adipose-derived mesenchymal stem cells were seeded on the hydrogel and the results of MTT assay, live/dead staining, and SEM images revealed that the scaffold with 1% dECM had better viscosity, cell viability, and proliferation. The study was conducted on the optimized scaffold (1% dECM) to determine mechanical characteristics, chondrogenic differentiation, and results demonstrated that the scaffold showed mechanical similarity to the native nasal cartilage tissue along with possessing appropriate biochemical features, which makes this new formulation based on PCL/dECM/Alg:Alg-Sul a promising candidate for further in-vivo studies.
Collapse
Affiliation(s)
- Pariya Zare
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | | | - Seyed Mohammad Davachi
- Department of Biology and Chemistry, Texas A&M International University, Laredo, Texas, USA
| | - Hadi Chahsetareh
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, Iran
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Negin Asgari
- Department of Biomedical Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Amin Haramshahi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rafieh Alizadeh
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Zohreh Bagher
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mohamad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
10
|
Li Y, Xun X, Xu Y, Zhan A, Gao E, Yu F, Wang Y, Luo H, Yang C. Hierarchical porous bacterial cellulose scaffolds with natural biomimetic nanofibrous structure and a cartilage tissue-specific microenvironment for cartilage regeneration and repair. Carbohydr Polym 2022; 276:118790. [PMID: 34823800 DOI: 10.1016/j.carbpol.2021.118790] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/10/2021] [Accepted: 10/16/2021] [Indexed: 12/20/2022]
Abstract
The limited three-dimensional (3D) nano-scale pore structure and lack of biological function hamper the application of bacterial cellulose (BC) in cartilage tissue engineering. To address this challenge, 3D hierarchical porous BC/decellularized cartilage extracellular matrix (DCECM) scaffolds with structurally and biochemically biomimetic cartilage regeneration microenvironment were fabricated by freeze-drying technique after EDC/NHS chemical crosslinking. The BC/DCECM scaffolds exhibited excellent mechanical properties, water superabsorbency and shape-memory properties. Compared with the BC control, the BC/DCECM scaffolds exhibited enhanced cell adhesion and proliferation. Cartilage regeneration in vitro and in vivo indicated that the BC/DCECM scaffolds achieved satisfactory neocartilage tissue regeneration with superior original shape fidelity, exterior natural cartilage-like appearance and histologically cartilage-specific lacuna formation and ECM deposition. Furthermore, the BC/DCECM scaffolds achieved superior repair outcomes, as hyaline cartilage-like tissue formed within the defect sites. The present study constitutes a strong step toward the further application of BC in cartilage tissue engineering.
Collapse
Affiliation(s)
- Yaqiang Li
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong middle Road, Shanghai 200001, China
| | - Xiaowei Xun
- Jiangxi Key Laboratory of Nanobiomaterials, Institute of Advanced Materials, East China Jiaotong University, Nanchang 330013, China
| | - Yong Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Anqi Zhan
- Institute of Plastic Surgery, Shandong Provincial Key Laboratory of Plastic and Microscopic Repair Technology, Weifang Medical University, Shandong 261053, China
| | - Erji Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Fan Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - You Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong middle Road, Shanghai 200001, China.
| | - Honglin Luo
- Jiangxi Key Laboratory of Nanobiomaterials, Institute of Advanced Materials, East China Jiaotong University, Nanchang 330013, China; School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China.
| | - Chunxi Yang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong middle Road, Shanghai 200001, China.
| |
Collapse
|
11
|
Levingstone TJ, Moran C, Almeida HV, Kelly DJ, O'Brien FJ. Layer-specific stem cell differentiation in tri-layered tissue engineering biomaterials: Towards development of a single-stage cell-based approach for osteochondral defect repair. Mater Today Bio 2021; 12:100173. [PMID: 34901823 PMCID: PMC8640516 DOI: 10.1016/j.mtbio.2021.100173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 12/26/2022] Open
Abstract
Successful repair of osteochondral defects is challenging, due in part to their complex gradient nature. Tissue engineering approaches have shown promise with the development of layered scaffolds that aim to promote cartilage and bone regeneration within the defect. The clinical potential of implanting these scaffolds cell-free has been demonstrated, whereby cells from the host bone marrow MSCs infiltrate the scaffolds and promote cartilage and bone regeneration within the required regions of the defect. However, seeding the cartilage layer of the scaffold with a chondrogenic cell population prior to implantation may enhance cartilage tissue regeneration, thus enabling the treatment of larger defects. Here the development of a cell seeding approach capable of enhancing articular cartilage repair without the requirement for in vitro expansion of the cell population is explored. The intrinsic ability of a tri-layered scaffold previously developed in our group to direct stem cell differentiation in each layer of the scaffold was first demonstrated. Following this, the optimal chondrogenic cell seeding approach capable of enhancing the regenerative capacity of the tri-layered scaffold was demonstrated with the highest levels of chondrogenesis achieved with a co-culture of rapidly isolated infrapatellar fat pad MSCs (FPMSCs) and chondrocytes (CCs). The addition of FPMSCs to a relatively small number of CCs led to a 7.8-fold increase in the sGAG production over chondrocytes in mono-culture. This cell seeding approach has the potential to be delivered within a single-stage approach, without the requirement for costly in vitro expansion of harvested cells, to achieve rapid repair of osteochondral defects. Tri-layered scaffold capable of directing layer specific stem cell differentiation. Potential of cell seeding regimes to enhance chondrogenic repair explored. Optimal cell seeding regime was an infrapatellar fat pad MSC:chondrocyte coculture. Adding infrapatellar fat pad MSCs to chondrocytes led to >7-fold increase in sGAG. This cell-seeded scaffold has potential for rapid repair of osteochondral defects.
Collapse
Affiliation(s)
- Tanya J. Levingstone
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen's Green, Dublin, 2, Ireland
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, 9, Ireland
- Centre for Medical Engineering Research (MEDeng), Dublin City University, Dublin, 9, Ireland
- Advanced Processing Technology Research Centre, Dublin City University, Dublin, 9, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, 2, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland
| | - Conor Moran
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen's Green, Dublin, 2, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, 2, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland
| | - Henrique V. Almeida
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, 2, Ireland
- iBET, Instituto de Biologia Experimental e Tecnológica, 2781-901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, 2, Ireland
| | - Daniel J. Kelly
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, 9, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, 2, Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen's Green, Dublin, 2, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, 2, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland
- Corresponding author. Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen's Green, Dublin, 2, Ireland.
| |
Collapse
|
12
|
Zhang X, Liu Y, Zuo Q, Wang Q, Li Z, Yan K, Yuan T, Zhang Y, Shen K, Xie R, Fan W. 3D Bioprinting of Biomimetic Bilayered Scaffold Consisting of Decellularized Extracellular Matrix and Silk Fibroin for Osteochondral Repair. Int J Bioprint 2021; 7:401. [PMID: 34825099 PMCID: PMC8611412 DOI: 10.18063/ijb.v7i4.401] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/20/2021] [Indexed: 12/27/2022] Open
Abstract
Recently, three-dimensional (3D) bioprinting technology is becoming an appealing approach for osteochondral repair. However, it is challenging to develop a bilayered scaffold with anisotropic structural properties to mimic a native osteochondral tissue. Herein, we developed a bioink consisting of decellularized extracellular matrix and silk fibroin to print the bilayered scaffold. The bilayered scaffold mimics the natural osteochondral tissue by controlling the composition, mechanical properties, and growth factor release in each layer of the scaffold. The in vitro results show that each layer of scaffolds had a suitable mechanical strength and degradation rate. Furthermore, the scaffolds encapsulating transforming growth factor-beta (TGF-β) and bone morphogenetic protein-2 (BMP-2) can act as a controlled release system and promote directed differentiation of bone marrow-derived mesenchymal stem cells. Furthermore, the in vivo experiments suggested that the scaffolds loaded with growth factors promoted osteochondral regeneration in the rabbit knee joint model. Consequently, the biomimetic bilayered scaffold loaded with TGF-β and BMP-2 would be a promising strategy for osteochondral repair.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang Zuo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qingyun Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zuxi Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kai Yan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Yuan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kai Shen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Xie
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weimin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Wang B, Díaz-Payno PJ, Browe DC, Freeman FE, Nulty J, Burdis R, Kelly DJ. Affinity-bound growth factor within sulfated interpenetrating network bioinks for bioprinting cartilaginous tissues. Acta Biomater 2021; 128:130-142. [PMID: 33866035 DOI: 10.1016/j.actbio.2021.04.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
3D bioprinting has emerged as a promising technology in the field of tissue engineering and regenerative medicine due to its ability to create anatomically complex tissue substitutes. However, it still remains challenging to develop bioactive bioinks that provide appropriate and permissive environments to instruct and guide the regenerative process in vitro and in vivo. In this study alginate sulfate, a sulfated glycosaminoglycan (sGAG) mimic, was used to functionalize an alginate-gelatin methacryloyl (GelMA) interpenetrating network (IPN) bioink to enable the bioprinting of cartilaginous tissues. The inclusion of alginate sulfate had a limited influence on the viscosity, shear-thinning and thixotropic properties of the IPN bioink, enabling high-fidelity bioprinting and supporting mesenchymal stem cell (MSC) viability post-printing. The stiffness of printed IPN constructs greatly exceeded that achieved by printing alginate or GelMA alone, while maintaining resilience and toughness. Furthermore, given the high affinity of alginate sulfate to heparin-binding growth factors, the sulfated IPN bioink supported the sustained release of transforming growth factor-β3 (TGF-β3), providing an environment that supported robust chondrogenesis in vitro, with little evidence of hypertrophy or mineralization over extended culture periods. Such bioprinted constructs also supported chondrogenesis in vivo, with the controlled release of TGF-β3 promoting significantly higher levels of cartilage-specific extracellular matrix deposition. Altogether, these results demonstrate the potential of bioprinting sulfated bioinks as part of a 'single-stage' or 'point-of-care' strategy for regenerating cartilaginous tissues. STATEMENT OF SIGNIFICANCE: This study highlights the potential of using sulfated interpenetrating network (IPN) bioink to support the regeneration of phenotypically stable articular cartilage. Construction of interpenetrating networks in the bioink enables unique high-fidelity bioprinting and provides synergistic increases in mechanical properties. The presence of alginate sulfate enables the capacity of high affinity-binding of TGF-β3, which promoted robust chondrogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Bin Wang
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Pedro J Díaz-Payno
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - David C Browe
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Fiona E Freeman
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Jessica Nulty
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Ross Burdis
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland; Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
14
|
Ravari MK, Mashayekhan S, Zarei F, Sayyahpour FA, Taghiyar L, Baghban Eslaminejad M. Fabrication and characterization of an injectable reinforced composite scaffold for cartilage tissue engineering: an in vitro study. Biomed Mater 2021; 16:045007. [PMID: 33784250 DOI: 10.1088/1748-605x/abed97] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There are limitations in current medications of articular cartilage injuries. Although injectable bioactive hydrogels are promising options, they have decreased biomechanical performance. Researchers should consider many factors when providing solutions to overcome these challenges. In this study, we created an injectable composite hydrogel from chitosan and human acellular cartilage extracellular matrix (ECM) particles. In order to enhance its mechanical properties, we reinforced this hydrogel with microporous microspheres composed of the same materials as the structural building blocks of the scaffold. Articular cartilage from human donors was decellularized by a combination of physical, chemical, and enzymatic methods. The decellularization efficiency was assessed by histological analysis and assessment of DNA content. We characterized the composite constructs in terms of storage modulus, gelation time, biocompatibility, and differentiation potential. The results showed that mechanical behavior increased with an increase in microsphere content. The sample that contained 10% microsphere had an enhanced storage modulus of up to 90 kPa. Biocompatibility and preliminary differentiation investigations revealed that this composite hydrogel might have potential benefits for cartilage tissue engineering.
Collapse
Affiliation(s)
- Mojtaba Khozaei Ravari
- Department of chemical and petroleum engineering, Sharif University of Technology, Tehran 11365-8639, Iran. Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | | | | | | | | | | |
Collapse
|
15
|
Khajavi M, Hajimoradloo A, Zandi M, Pezeshki-Modaress M, Bonakdar S, Zamani A. Fish cartilage: A promising source of biomaterial for biological scaffold fabrication in cartilage tissue engineering. J Biomed Mater Res A 2021; 109:1737-1750. [PMID: 33738960 DOI: 10.1002/jbm.a.37169] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/17/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022]
Abstract
Here, engineered cartilage-like scaffold using an extracellular matrix (ECM) from sturgeon fish cartilage provided a chondroinductive environment to stimulate cartilaginous matrix synthesis in human adipose stem cells (hASCs). Three dimensional porous and degradable fish cartilage ECM-derived scaffold (FCS) was produced using a protocol containing chemical decellularization, enzymatic solubilization, freeze-drying and EDC-crosslinking treatments and the effect of different ECM concentrations (10, 20, 30, and 40 mg/ml) on prepared scaffolds was investigated through physical, mechanical and biological analysis. The histological and scanning electron microscopy analysis revealed the elimination of the cell fragments and a 3-D interconnected porous structure, respectively. Cell viability assay displayed no cytotoxic effects. The prepared porous constructs of fish cartilage ECM were seeded with hASCs for 21 days and compared to collagen (Col) and collagen-10% hyaluronic acid (Col-HA) scaffolds. Cell culture results evidenced that the fabricated scaffolds could provide a proper 3-D structure to support the adhesion, proliferation and chondrogenic differentiation of hASCs considering the synthesis of specific proteins of cartilage, collagen type II (Col II) and aggrecan (ACAN). Based on the results of the present study, it can be concluded that the porous scaffold derived from fish cartilage ECM possesses an excellent potential for cartilage tissue engineering.
Collapse
Affiliation(s)
- Maryam Khajavi
- Department of Fisheries, Faculty of Fisheries and Environmental Science, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Abdolmajid Hajimoradloo
- Department of Fisheries, Faculty of Fisheries and Environmental Science, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Mojgan Zandi
- Department of Biomaterials, Iran Polymer and Petrochemical Institute, Tehran, Iran
| | | | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Abbas Zamani
- Department of Fisheries, Faculty of Natural Resources and Environment, Malayer University, Malayer, Iran
| |
Collapse
|
16
|
Matheson AR, Sheehy EJ, Jay GD, Scott WM, O'Brien FJ, Schmidt TA. The role of synovial fluid constituents in the lubrication of collagen-glycosaminoglycan scaffolds for cartilage repair. J Mech Behav Biomed Mater 2021; 118:104445. [PMID: 33740688 DOI: 10.1016/j.jmbbm.2021.104445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 11/18/2022]
Abstract
Extracellular matrix (ECM)-derived scaffolds have shown promise as tissue-engineered grafts for promoting cartilage repair. However, there has been a lack of focus on fine-tuning the frictional properties of scaffolds for cartilage tissue engineering as well as understanding their interactions with synovial fluid constituents. Proteoglycan-4 (PRG4) and hyaluronan (HA) are macromolecules within synovial fluid that play key roles as boundary mode lubricants during cartilage surface interactions. The overall objective of this study was to characterize the role PRG4 and HA play in the lubricating function of collagen-glycosaminoglycan (GAG) scaffolds for cartilage repair. As a first step towards this goal, we aimed to develop a suitable in vitro friction test to establish the boundary mode lubrication parameters for collagen-GAG scaffolds articulated against glass in a phosphate buffered saline (PBS) bath. Subsequently, we sought to leverage this system to determine the effect of physiological synovial fluid lubricants, PRG4 and HA, on the frictional properties of collagen-GAG scaffolds, with scaffolds hydrated in PBS and bovine synovial fluid (bSF) serving as negative and positive controls, respectively. At all compressive strains examined (ε = 0.1-0.5), fluid depressurization within hydrated collagen-GAG scaffolds was >99% complete at ½ minute. The coefficient of friction was stable at all compressive strains (ranging from a low 0.103 ± 0.010 at ε = 0.3 up to 0.121 ± 0.015 at ε = 0.4) and indicative of boundary-mode conditions. Immunohistochemistry demonstrated that PRG4 from recombinant human (rh) and bovine sources adsorbed to collagen-GAG scaffolds and the coefficient of friction for scaffolds immersed in rhPRG4 (0.067 ± 0.027) and normal bSF (0.056 ± 0.020) solution decreased compared to PBS (0.118 ± 0.21, both p < 0.05, at ε = 0.2). The ability of the adsorbed rhPRG4 to reduce friction on the scaffolds indicates that its incorporation within collagen-GAG biomaterials may enhance their lubricating ability as potential tissue-engineered cartilage replacements. To conclude, this study reports the development of an in vitro friction test capable of characterizing the coefficient of friction of ECM-derived scaffolds tested in a range of synovial fluid lubricants and demonstrates frictional properties as a potential design parameter for implants and materials for soft tissue replacement.
Collapse
Affiliation(s)
- Austyn R Matheson
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Eamon J Sheehy
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Gregory D Jay
- Department of Emergency Medicine, Warren Alpert Medical School & School of Engineering, Brown University, Providence, RI, USA
| | - W Michael Scott
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada; Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Fergal J O'Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Tannin A Schmidt
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
17
|
Zhou L, Gjvm VO, Malda J, Stoddart MJ, Lai Y, Richards RG, Ki-Wai Ho K, Qin L. Innovative Tissue-Engineered Strategies for Osteochondral Defect Repair and Regeneration: Current Progress and Challenges. Adv Healthc Mater 2020; 9:e2001008. [PMID: 33103381 DOI: 10.1002/adhm.202001008] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/19/2020] [Indexed: 12/20/2022]
Abstract
Clinical treatments for the repair of osteochondral defects (OCD) are merely palliative, not completely curative, and thus enormously unfulfilled challenges. With the in-depth studies of biology, medicine, materials, and engineering technology, the conception of OCD repair and regeneration should be renewed. During the past decades, many innovative tissue-engineered approaches for repairing and regenerating damaged osteochondral units have been widely explored. Various scaffold-free and scaffold-based strategies, such as monophasic, biphasic, and currently fabricated multiphasic and gradient architectures have been proposed and evaluated. Meanwhile, progenitor cells and tissue-specific cells have also been intensively investigated in vivo as well as ex vivo. Concerning bioactive factors and drugs, they have been combined with scaffolds and/or living cells, and even released in a spatiotemporally controlled manner. Although tremendous progress has been achieved, further research and development (R&D) is needed to convert preclinical outcomes into clinical applications. Here, the osteochondral unit structure, its defect classifications, and diagnosis are summarized. Commonly used clinical reparative techniques, tissue-engineered strategies, emerging 3D-bioprinting technologies, and the status of their clinical applications are discussed. Existing challenges to translation are also discussed and potential solutions for future R&D directions are proposed.
Collapse
Affiliation(s)
- Liangbin Zhou
- Musculoskeletal Research Laboratory of Department of Orthopedics & Traumatology, and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Van Osch Gjvm
- Department of Orthopedics and Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Rotterdam, 3000 CA, The Netherlands
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Delft, 2600 AA, The Netherlands
| | - Jos Malda
- Department of Orthopaedics of University Medical Center Utrecht, and Department of Clinical Sciences of Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CS, The Netherlands
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, Davos, CH 7270, Switzerland
| | - Yuxiao Lai
- Centre for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, The Chinese Academy of Sciences, Shenzhen, 518000, China
| | - R Geoff Richards
- AO Research Institute Davos, Clavadelerstrasse 8, Davos, CH 7270, Switzerland
| | - Kevin Ki-Wai Ho
- Musculoskeletal Research Laboratory of Department of Orthopedics & Traumatology, and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopedics & Traumatology, and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
- Centre for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, The Chinese Academy of Sciences, Shenzhen, 518000, China
| |
Collapse
|
18
|
Zhang X, Liu Y, Luo C, Zhai C, Li Z, Zhang Y, Yuan T, Dong S, Zhang J, Fan W. Crosslinker-free silk/decellularized extracellular matrix porous bioink for 3D bioprinting-based cartilage tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111388. [PMID: 33254994 DOI: 10.1016/j.msec.2020.111388] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/14/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
As cartilage tissue lacks the innate ability to mount an adequate regeneration response, damage to it is detrimental to the quality of life of the subject. The emergence of three-dimensional bioprinting (3DBP) technology presents an opportunity to repair articular cartilage defects. However, widespread adoption of this technique has been impeded by difficulty in preparing a suitable bioink and the toxicity inherent in the chemical crosslinking process of most bioinks. Our objective was to develop a crosslinker-free bioink with the same biological activity as the original cartilage extracellular matrix (ECM) and good mechanical strength. We prepared bioinks containing different concentrations of silk fibroin and decellularized extracellular matrix (SF-dECM bioinks) mixed with bone marrow mesenchymal stem cells (BMSCs) for 3D bioprinting. SF and dECM interconnect with each other through physical crosslinking and entanglement. A porous structure was formed by removing the polyethylene glycol from the SF-dECM bioink. The results showed the SF-dECM construct had a suitable mechanical strength and degradation rate, and the expression of chondrogenesis-specific genes was found to be higher than that of the SF control construct group. Finally, we confirmed that a SF-dECM construct that was designed to release TGF-β3 had the ability to promote chondrogenic differentiation of BMSCs and provided a good cartilage repair environment, suggesting it is an ideal scaffold for cartilage tissue engineering.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Chunyang Luo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Chenjun Zhai
- Department of Orthopedics, Yixing People's Hospital, Yixing, Jiangsu 214200, China
| | - Zuxi Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Tao Yuan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Shilei Dong
- Key Lab of Biofabrication of AnHui Higher Education Institution Centre for Advanced Biofabrication, Hefei, Anhui 230601, China
| | - Jiyong Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Weimin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
19
|
Greif DN, Kouroupis D, Murdock CJ, Griswold AJ, Kaplan LD, Best TM, Correa D. Infrapatellar Fat Pad/Synovium Complex in Early-Stage Knee Osteoarthritis: Potential New Target and Source of Therapeutic Mesenchymal Stem/Stromal Cells. Front Bioeng Biotechnol 2020; 8:860. [PMID: 32850724 PMCID: PMC7399076 DOI: 10.3389/fbioe.2020.00860] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022] Open
Abstract
The infrapatellar fat pad (IFP) has until recently been viewed as a densely vascular and innervated intracapsular/extrasynovial tissue with biomechanical roles in the anterior compartment of the knee. Over the last decade, secondary to the proposition that the IFP and synovium function as a single unit, its recognized tight molecular crosstalk with emerging roles in the pathophysiology of joint disease, and the characterization of immune-related resident cells with varying phenotypes (e.g., pro and anti-inflammatory macrophages), this structural complex has gained increasing attention as a potential therapeutic target in patients with various knee pathologies including osteoarthritis (KOA). Furthermore, the description of the presence of mesenchymal stem/stromal cells (MSC) as perivascular cells within the IFP (IFP-MSC), exhibiting immunomodulatory, anti-fibrotic and neutralizing activities over key local mediators, has promoted the IFP as an alternative source of MSC for cell-based therapy protocols. These complementary concepts have supported the growing notion of immune and inflammatory events participating in the pathogenesis of KOA, with the IFP/synovium complex engaging not only in amplifying local pathological responses, but also as a reservoir of potential therapeutic cell-based products. Consequently, the aim of this review is to outline the latest discoveries related with the IFP/synovium complex as both an active participant during KOA initiation and progression thus emerging as a potential target, and a source of therapeutic IFP-MSCs. Finally, we discuss how these notions may help the design of novel treatments for KOA through modulation of local cellular and molecular cascades that ultimately lead to joint destruction.
Collapse
Affiliation(s)
- Dylan N Greif
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Christopher J Murdock
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Lee D Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
20
|
Zhao Y, Teng B, Sun X, Dong Y, Wang S, Hu Y, Wang Z, Ma X, Yang Q. Synergistic Effects of Kartogenin and Transforming Growth Factor-β3 on Chondrogenesis of Human Umbilical Cord Mesenchymal Stem Cells In Vitro. Orthop Surg 2020; 12:938-945. [PMID: 32462800 PMCID: PMC7307229 DOI: 10.1111/os.12691] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/29/2020] [Accepted: 04/03/2020] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE To explore the effect of kartogenin (KGN) on proliferation and chondrogenic differentiation of human umbilical cord mesenchymal stem cells (hUCMSC) in vitro, and the synergistic effects of KGN and transforming growth factor (TGF)-β3 on hUCMSC. METHODS Human umbilical cord mesenchymal stem cells were isolated and cultured. Then the differentiation properties were identified by flow cytometry analysis. HUCMSC were divided into four groups: control group, KGN group, TGF-β3 group, and TK group (with TGF-β3 and KGN added into the medium simultaneously). Cells in all groups were induced for 21 days using the suspension ball culture method. Hematoxylin and eosin, immunofluorescence, and Alcian blue staining were used to analyze chondrogenic differentiation. Real-time reverse transcriptase polymerase chain reaction was performed to investigate genes associated with chondrogenic differentiation. RESULT Hematoxylin and eosin staining showed that cells in the TGF-β3 group and the TK group had formed cartilage-like tissue after 21 days of culture. The results of immunofluorescence and Alcian blue staining showed that compared with the control group, cells in the KGN and TGF-β3 groups demonstrated increased secretion of aggrecan after 21 days of culture. In addition, cells in the group combining KGN with TGF-β3 (5.587 ± 0.27, P < 0.01) had more collagen II secretion than cells in the TGF-β3 alone group (2.86 ± 0.141, P < 0.01) or the KGN group (1.203 ± 0.215, P < 0.01). The expression of aggrecan (2.468 ± 0.097, P < 0.05) and SRY-Box 9 (4.08 ± 0.13, P < 0.05) in cells in the group combining KGN with TGF-β3 was significantly higher than those in the TGF-β3 group (2.216 ± 0.09, 3.02 ± 0.132, P < 0.05).' CONCLUSION The combination of KGN and TGF-β3 had synergistic effects and induced hUCMSC chondrogenesis. This could represent a new approach for clinical application and studies on cartilage repair and regeneration.
Collapse
Affiliation(s)
- Yanhong Zhao
- Stomatological Hospital of Tianjin Medical UniversityTianjinChina
| | - Binhong Teng
- Stomatological Hospital of Tianjin Medical UniversityTianjinChina
- Department of Oral and Maxillofacial SurgerySchool and Hospital of Stomatology, Peking UniversityBeijingChina
| | - Xun Sun
- Department of Spine SurgeryTianjin Hospital, Tianjin UniversityTianjinChina
| | - Yunsheng Dong
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai UniversityTianjinChina
| | - Shufang Wang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai UniversityTianjinChina
| | - Yongcheng Hu
- Department of Spine SurgeryTianjin Hospital, Tianjin UniversityTianjinChina
| | - Zheng Wang
- Department of OrthopedicsNo. 1 Medical Center of Chinese PLA General HospitalBeijingChina
| | - Xinlong Ma
- Department of Spine SurgeryTianjin Hospital, Tianjin UniversityTianjinChina
| | - Qiang Yang
- Department of Spine SurgeryTianjin Hospital, Tianjin UniversityTianjinChina
| |
Collapse
|
21
|
Díaz-Payno PJ, Browe DC, Cunniffe GM, Kelly DJ. The identification of articular cartilage and growth plate extracellular matrix-specific proteins supportive of either osteogenesis or stable chondrogenesis of stem cells. Biochem Biophys Res Commun 2020; 528:285-291. [PMID: 32473752 DOI: 10.1016/j.bbrc.2020.05.074] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Tissue-specific extracellular matrix (ECM) proteins can play a key role in regulating the fate of stem cells and can potentially be utilized for therapeutic applications. Realising this potential requires further characterization of the diversity of biomolecules present in tissue-specific ECMs and an evaluation of their role as regulatory cues for regenerative medicine applications. The goal of this study was to identify specific soluble factors within the ECM of articular cartilage (AC) and growth plate (GP) that may impart chondro-inductivity or osteo-inductivity respectively. To this end, the significantly different proteins between both matrisomes were searched against the STRING database platform, from which C-type lectin domain family-11 member-A (CLEC11A) and S100 calcium-binding protein-A10 (S100A10) were identified as potential candidates for supporting osteogenesis, and Gremlin-1 (GREM1) and TGF-β induced gene human clone-3 (βIGH3) were identified as potential candidates for supporting stable chondrogenesis. Stimulation of chondrogenically-primed bone marrow-derived stem cells (BMSCs) with the AC-specific proteins GREM1 and βIGH3 had no noticeable effect on the deposition of collagen-II, a marker of chondrogenesis, but appeared to suppress the production of the hypertrophic marker collagen-X, particularly for higher concentrations of GREM1. Stimulation with GREM1 was also found to suppress the direct osteoblastic differentiation of BMSCs. In contrast, stimulation with the GP-specific factors CLEC11A and S100A10 was found to enhance osteogenesis of BMSCs, increasing the levels of mineralization, particularly for higher concentration of CLEC11A. Together these results demonstrate that AC- and GP-specific proteins may play a key role in developing novel strategies for engineering phenotypically stable articular cartilage or enhancing the regeneration of critically-sized bone defects.
Collapse
Affiliation(s)
- Pedro J Díaz-Payno
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - David C Browe
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Gráinne M Cunniffe
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland; Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
22
|
Liu Q, Li XJ, Wang ZS. [Effects of porcine acellular cartilaginous matrix on the proliferation and differentiation of human adipose-derived stromal cells]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:122-127. [PMID: 32314882 DOI: 10.7518/hxkq.2020.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE This study aimed to evaluate the effects of porcine acellular cartilaginous matrix (pACM) on the proliferation and differentiation of human adipose-derived stromal cells (hADSCs). METHODS pACM was prepared from porcine articular cartilage through decellularization treatment. hADSCs were isolated from human adipose tissues and cultured with different pACM concentrations. No pACM was used as the control group. The effect of pACM on hADSCs proliferation was detected by CCK-8 method. Moreover, the effect of pACM on hADSCs chondrogenic differentiation was analyzed through fluorescence quantitative polymerase chain reaction and Western blot. RESULTS hADSCs proliferation rate in 0.5, 1.0, and 2.0 mg·mL⁻¹ pACM groups was not significantly different from that in the control group, whereas that in 4.0 and 8.0 mg·mL⁻¹ pACM group was lower than that in the control group (P<0.05). The expression levels of pACM chondrogenic genes, including SOX-9, collagen type Ⅱ alpha 1 chain (COL2A1), and aggrecan (ACAN) and cell adhesion-related gene LAMININ in 0.5, 1.0, and 2.0 mg·mL⁻¹ pACM group were higher than those of the control group (P<0.05), but that of a stemness-related gene Notch-1 was lower than that of the control group (P<0.05). No statistical difference was found in the expression of a lipogenesis-related gene peroxisome proliferator-activated receptor-γ (PPAr-γ) (P>0.05). The expression levels of chondrogenic proteins (SOX-9, COL2A1, and ACAN) were higher than those of the control group (P<0.05). CONCLUSIONS Appropriate pACM con-centrations do not affect hADSCs proliferation but can induce hADSCs chondrogenic differentiation.
Collapse
Affiliation(s)
- Qian Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology, Dept. of Prosthodontics, School of Stomatology, Air Force Medical University, Xi'an 710032, China
| | - Xue-Jian Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology, Dept. of Prosthodontics, School of Stomatology, Air Force Medical University, Xi'an 710032, China
| | - Zhong-Shan Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology, Dept. of Prosthodontics, School of Stomatology, Air Force Medical University, Xi'an 710032, China
| |
Collapse
|
23
|
Zhao Y, Zhao X, Zhang R, Huang Y, Li Y, Shan M, Zhong X, Xing Y, Wang M, Zhang Y, Zhao Y. Cartilage Extracellular Matrix Scaffold With Kartogenin-Encapsulated PLGA Microspheres for Cartilage Regeneration. Front Bioeng Biotechnol 2020; 8:600103. [PMID: 33363129 PMCID: PMC7756004 DOI: 10.3389/fbioe.2020.600103] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Repair of articular cartilage defects is a challenging aspect of clinical treatment. Kartogenin (KGN), a small molecular compound, can induce the differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) into chondrocytes. Here, we constructed a scaffold based on chondrocyte extracellular matrix (CECM) and poly(lactic-co-glycolic acid) (PLGA) microspheres (MP), which can slowly release KGN, thus enhancing its efficiency. Cell adhesion, live/dead staining, and CCK-8 results indicated that the PLGA(KGN)/CECM scaffold exhibited good biocompatibility. Histological staining and quantitative analysis demonstrated the ability of the PLGA(KGN)/CECM composite scaffold to promote the differentiation of BMSCs. Macroscopic observations, histological tests, and specific marker analysis showed that the regenerated tissues possessed characteristics similar to those of normal hyaline cartilage in a rabbit model. Use of the PLGA(KGN)/CECM scaffold may mimic the regenerative microenvironment, thereby promoting chondrogenic differentiation of BMSCs in vitro and in vivo. Therefore, this innovative composite scaffold may represent a promising approach for acellular cartilage tissue engineering.
Collapse
Affiliation(s)
- Yanhong Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
- *Correspondence: Yanhong Zhao,
| | - Xige Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Rui Zhang
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Ying Huang
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Yunjie Li
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Minhui Shan
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Xintong Zhong
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Yi Xing
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Min Wang
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | | | - Yanmei Zhao
- Institute of Disaster Medicine, Tianjin University, Tianjin, China
- Yanmei Zhao,
| |
Collapse
|
24
|
Daly AC, Riley L, Segura T, Burdick JA. Hydrogel microparticles for biomedical applications. NATURE REVIEWS. MATERIALS 2020; 5:20-43. [PMID: 34123409 PMCID: PMC8191408 DOI: 10.1038/s41578-019-0148-6] [Citation(s) in RCA: 524] [Impact Index Per Article: 131.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/18/2019] [Indexed: 05/13/2023]
Abstract
Hydrogel microparticles (HMPs) are promising for biomedical applications, ranging from the therapeutic delivery of cells and drugs to the production of scaffolds for tissue repair and bioinks for 3D printing. Biologics (cells and drugs) can be encapsulated into HMPs of predefined shapes and sizes using a variety of fabrication techniques (batch emulsion, microfluidics, lithography, electrohydrodynamic (EHD) spraying and mechanical fragmentation). HMPs can be formulated in suspensions to deliver therapeutics, as aggregates of particles (granular hydrogels) to form microporous scaffolds that promote cell infiltration or embedded within a bulk hydrogel to obtain multiscale behaviours. HMP suspensions and granular hydrogels can be injected for minimally invasive delivery of biologics, and they exhibit modular properties when comprised of mixtures of distinct HMP populations. In this Review, we discuss the fabrication techniques that are available for fabricating HMPs, as well as the multiscale behaviours of HMP systems and their functional properties, highlighting their advantages over traditional bulk hydrogels. Furthermore, we discuss applications of HMPs in the fields of cell delivery, drug delivery, scaffold design and biofabrication.
Collapse
Affiliation(s)
- Andrew C. Daly
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- These authors contributed equally: Andrew C. Daly, Lindsay Riley
| | - Lindsay Riley
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- These authors contributed equally: Andrew C. Daly, Lindsay Riley
| | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Departments of Dermatology and Neurology, Duke University, Durham, NC, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Beigi MH, Atefi A, Ghanaei HR, Labbaf S, Ejeian F, Nasr-Esfahani MH. Activated platelet-rich plasma improves cartilage regeneration using adipose stem cells encapsulated in a 3D alginate scaffold. J Tissue Eng Regen Med 2019. [PMID: 29522657 DOI: 10.1002/term.2663] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In the current study, the effect of superimposing platelet-rich plasma (PRP) on different culture mediums in a three-dimensional alginate scaffold encapsulated with adipose-derived mesenchymal stem cells for cartilage tissue repair is reported. The three-dimensional alginate scaffolds with co-administration of PRP and/or chondrogenic supplements had a significant effect on the differentiation of adipose mesenchymal stem cells into mature cartilage, as assessed by an evaluation of the expression of cartilage-related markers of Sox9, collagen II, aggrecan and collagen, and glycosaminoglycan assays. For in vivo studies, following induction of osteochondral lesion in a rabbit model, a high degree of tissue regeneration in the alginate plus cell group (treated with PRP plus chondrogenic medium) compared with other groups of cell-free alginate and untreated groups (control) were observed. After 8 weeks, in the alginate plus cell group, functional chondrocytes were observed, which produced immature matrix, and by 16 weeks, the matrix and hyaline-like cartilage became completely homogeneous and integrated with the natural surrounding cartilage in the defect site. Similar effect was also observed in the subchondral bone. The cell-free scaffolds formed fibrocartilage tissue, and the untreated group did not form a continuous cartilage over the defect by 16 weeks.
Collapse
Affiliation(s)
- Mohammad-Hossein Beigi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Atefeh Atefi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hamid-Reza Ghanaei
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sheyda Labbaf
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | - Fatemeh Ejeian
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad-Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
26
|
Transforming Growth Factor Beta 3-Loaded Decellularized Equine Tendon Matrix for Orthopedic Tissue Engineering. Int J Mol Sci 2019; 20:ijms20215474. [PMID: 31684150 PMCID: PMC6862173 DOI: 10.3390/ijms20215474] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/25/2019] [Accepted: 11/01/2019] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor beta 3 (TGFβ3) promotes tenogenic differentiation and may enhance tendon regeneration in vivo. This study aimed to apply TGFβ3 absorbed in decellularized equine superficial digital flexor tendon scaffolds, and to investigate the bioactivity of scaffold-associated TGFβ3 in an in vitro model. TGFβ3 could effectively be loaded onto tendon scaffolds so that at least 88% of the applied TGFβ3 were not detected in the rinsing fluid of the TGFβ3-loaded scaffolds. Equine adipose tissue-derived multipotent mesenchymal stromal cells (MSC) were then seeded on scaffolds loaded with 300 ng TGFβ3 to assess its bioactivity. Both scaffold-associated TGFβ3 and TGFβ3 dissolved in the cell culture medium, the latter serving as control group, promoted elongation of cell shapes and scaffold contraction (p < 0.05). Furthermore, scaffold-associated and dissolved TGFβ3 affected MSC musculoskeletal gene expression in a similar manner, with an upregulation of tenascin c and downregulation of other matrix molecules, most markedly decorin (p < 0.05). These results demonstrate that the bioactivity of scaffold-associated TGFβ3 is preserved, thus TGFβ3 application via absorption in decellularized tendon scaffolds is a feasible approach.
Collapse
|
27
|
Yang D, Xiao J, Wang B, Li L, Kong X, Liao J. The immune reaction and degradation fate of scaffold in cartilage/bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109927. [DOI: 10.1016/j.msec.2019.109927] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 01/05/2023]
|
28
|
Li Y, Liu Y, Xun X, Zhang W, Xu Y, Gu D. Three-Dimensional Porous Scaffolds with Biomimetic Microarchitecture and Bioactivity for Cartilage Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2019; 11:36359-36370. [PMID: 31509372 DOI: 10.1021/acsami.9b12206] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Ideal tissue-engineering cartilage scaffolds should possess the same nanofibrous structure as the microstructure of native cartilage as well as the same biological function provided by the microenvironment for neocartilage regeneration. In the present study, three-dimensional composite biomimetic scaffolds with different concentration ratios of electrospun gelatin-polycaprolactone (gelatin-PCL) nanofibers and decellularized cartilage extracellular matrix (DCECM) were fabricated. The nanofibers with the biomimetic microarchitecture of native cartilage served as a skeleton with excellent mechanical properties, and the DCECM served as a biological functionalization platform for the induction of cell response and the promotion of cartilage regeneration. Experimental results showed that the composite nanofiber/DCECM (NF/DCECM) scaffolds had stronger mechanical properties and structural stability in wet state compared with those of DCECM scaffolds. In vitro experiments demonstrated that all scaffolds had good biocompatibility, but the chondrocyte proliferation rate of the composite NF/DCECM scaffolds was higher than that of the NF scaffolds. In vitro and in vivo cartilage regeneration results indicated that the DCECM component of the composite scaffolds facilitated early maturation of the cartilage lacuna and the secretion of collagen and glycosaminoglycan. The macroscopic and histological results at 12 weeks postsurgery exhibited that the composite NF/DCECM scaffolds yielded better cartilage repair outcomes than those of the nontreated group and NF scaffolds group. Overall, the present study demonstrated that the structurally and functionally biomimetic NF/DCECM scaffold is a promising tissue engineering scaffold for cartilage regeneration and cartilage defect repair.
Collapse
Affiliation(s)
| | | | - Xiaowei Xun
- Institute of Advanced Materials , East China Jiaotong University , Nanchang 330013 , China
| | - Wei Zhang
- Institute of Plastic Surgery, Shandong Provincial Key Laboratory of Plastic and Microscopic Repair Technology , Weifang Medical University , Weifang , Shandong 261041 , China
| | - Yong Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital , Tongji University School of Medicine , Shanghai 200433 , China
| | | |
Collapse
|
29
|
Gonzalez-Fernandez T, Sikorski P, Leach JK. Bio-instructive materials for musculoskeletal regeneration. Acta Biomater 2019; 96:20-34. [PMID: 31302298 PMCID: PMC6717669 DOI: 10.1016/j.actbio.2019.07.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 06/26/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023]
Abstract
The prevalence and cost of disorders affecting the musculoskeletal system are predicted to rise significantly in the coming years due to the aging global population and the increase of associated risk factors. Despite being the second largest cause of disability, the clinical options for therapeutic intervention remain limited. The clinical translation of cell-based therapies for the treatment of musculoskeletal disorders faces many challenges including maintenance of cell survival in the harsh in vivo environment and the lack of control over regulating cell phenotype upon implantation. In order to address these challenges, the development of bio-instructive materials to modulate cell behavior has taken center stage as a strategy to increase the therapeutic potential of various cell populations. However, the determination of the necessary cues for a specific application and how these signals should be presented from a biomaterial remains elusive. This review highlights recent biochemical and physical strategies used to engineer bio-instructive materials for the repair of musculoskeletal tissues. There is a particular emphasis on emerging efforts such as the engineering of immunomodulatory and antibacterial materials, as well as the incorporation of these strategies into biofabrication and organ-on-a-chip approaches. STATEMENT OF SIGNIFICANCE: Disorders affecting the musculoskeletal system affect individuals across the lifespan and have a profound effect on mobility and quality of life. While small defects in many tissues can heal successfully, larger defects are often unable to heal or instead heal with inferior quality fibrous tissue and require clinical intervention. Cell-based therapies are a promising option for clinical translation, yet challenges related to maintaining cell survival and instructing cell phenotype upon implantation have limited the success of this approach. Bio-instructive materials provide an exciting opportunity to modulate cell behavior and enhance the efficacy of cell-based approaches for musculoskeletal repair. However, the identification of critical instructive cues and how to present these stimuli is a focus of intense investigation. This review highlights recent biochemical and physical strategies used to engineer bio-instructive materials for the repair of musculoskeletal tissues, while also considering exciting progress in the engineering of immunomodulatory and antibacterial materials.
Collapse
Affiliation(s)
| | - Pawel Sikorski
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Department of Physics, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA.
| |
Collapse
|
30
|
Patel DK, Lim KT. Biomimetic Polymer-Based Engineered Scaffolds for Improved Stem Cell Function. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2950. [PMID: 31514460 PMCID: PMC6766224 DOI: 10.3390/ma12182950] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022]
Abstract
Scaffolds are considered promising materials for tissue engineering applications due to their unique physiochemical properties. The high porosity and adequate mechanical properties of the scaffolds facilitate greater cell adhesion, proliferation, and differentiation. Stem cells are frequently applied in tissue engineering applications due to their excellent potential. It has been noted that cell functions are profoundly affected by the nature of the extracellular matrix (ECM). Naturally derived ECM contains the bioactive motif that also influences the immune response of the organism. The properties of polymer scaffolds mean they can resemble the native ECM and can regulate cellular responses. Various techniques such as electrospinning and 3D printing, among others, are frequently used to fabricate polymer scaffolds, and their cellular responses are different with each technique. Furthermore, enhanced cell viability, as well as the differentiation ability of stem cells on the surface of scaffolds, opens a fascinating approach to the formation of ECM-like environments for tissue engineering applications.
Collapse
Affiliation(s)
- Dinesh K Patel
- The Institute of Forest Science, Kangwon National University, Chuncheon-24341, Korea.
| | - Ki-Taek Lim
- Department of Biosystems Engineering, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon-24341, Korea.
| |
Collapse
|
31
|
The Role of Extracellular Matrix Expression, ERK1/2 Signaling and Cell Cohesiveness for Cartilage Yield from iPSCs. Int J Mol Sci 2019; 20:ijms20174295. [PMID: 31480758 PMCID: PMC6747490 DOI: 10.3390/ijms20174295] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023] Open
Abstract
Current therapies involving chondrocytes or mesenchymal stromal cells (MSCs) remain inefficient in restoring cartilage properties upon injury. The induced pluripotent stem-cell (iPSC)-derived mesenchymal progenitor cells (iMPCs) have been put forward as a promising alternative cell source due to their high proliferation and differentiation potential. However, the observed cell loss during in vitro chondrogenesis is currently a bottleneck in establishing articular chondrocyte generation from iPSCs. In a search for candidate mechanisms underlying the low iPSC-derived cartilage tissue yield, global transcriptomes were compared between iMPCs and MSCs and the cell properties were analyzed via a condensation assay. The iMPCs had a more juvenile mesenchymal gene signature than MSCs with less myofibroblast-like characteristics, including significantly lower ECM- and integrin-ligand-related as well as lower α-smooth-muscle-actin expression. This correlated with less substrate and more cell-cell adhesion, impaired aggregate formation and consequently inferior cohesive tissue properties of the iMPC-pellets. Along lower expression of pro-survival ECM molecules, like decorin, collagen VI, lumican and laminin, the iMPC populations had significantly less active ERK1/2 compared to MSCs. Overall, this study proposes that this ECM and integrin-ligand shortage, together with insufficient pro-survival ERK1/2-activity, explains the loss of a non-aggregating iMPC sub-fraction during pellet formation and reduced survival of cells in early pellets. Enhancing ECM production and related signaling in iMPCs may be a promising new means to enrich the instructive microenvironment with pro-survival cues allowing to improve the final cartilage tissue yield from iPSCs.
Collapse
|
32
|
Jiang R, Wang G, Zhang J, Zhang X, Zhou L, Xu T. Three-dimensional bioprinting of auricular cartilage: A review. MEDICINE IN DRUG DISCOVERY 2019. [DOI: 10.1016/j.medidd.2020.100016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
33
|
Zhai C, Zhang X, Chen J, He J, Fei H, Liu Y, Luo C, Fan W. The effect of cartilage extracellular matrix particle size on the chondrogenic differentiation of bone marrow mesenchymal stem cells. Regen Med 2019; 14:663-680. [PMID: 31313645 DOI: 10.2217/rme-2018-0082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Aim: To investigate the effect of cartilage extracellular matrix (ECM) particle size on the chondrogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Materials & methods: BMSCs were seeded into the scaffolds fabricated by small particle ECM materials and large particle ECM materials. For the positive control, chondrogenically induced BMSCs were seeded into commercial poly-lactic-glycolic acid scaffolds. Macroscopic observation, histological and immunohistochemical staining, mechanical testing and biochemical analysis were performed to the cell-scaffold constructs. Results: BMSCs in small particle ECM materials and poly-lactic-glycolic acid scaffolds were induced to differentiate into chondrocytes, while BMSCs in the large particle ECM materials scaffold did not differentiate into chondrocytes. Conclusion: The small ECM particle materials improved the induction ability of the cartilage ECM-derived scaffold.
Collapse
Affiliation(s)
- Chenjun Zhai
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Department of Orthopedics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214200, China
| | - Xiao Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jun Chen
- Department of Orthopedics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214200, China
| | - Jian He
- Department of Orthopedics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214200, China
| | - Hao Fei
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Chunyang Luo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Weimin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
34
|
Patel JM, Saleh KS, Burdick JA, Mauck RL. Bioactive factors for cartilage repair and regeneration: Improving delivery, retention, and activity. Acta Biomater 2019; 93:222-238. [PMID: 30711660 PMCID: PMC6616001 DOI: 10.1016/j.actbio.2019.01.061] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/25/2019] [Accepted: 01/29/2019] [Indexed: 12/29/2022]
Abstract
Articular cartilage is a remarkable tissue whose sophisticated composition and architecture allow it to withstand complex stresses within the joint. Once injured, cartilage lacks the capacity to self-repair, and injuries often progress to joint wide osteoarthritis (OA) resulting in debilitating pain and loss of mobility. Current palliative and surgical management provides short-term symptom relief, but almost always progresses to further deterioration in the long term. A number of bioactive factors, including drugs, corticosteroids, and growth factors, have been utilized in the clinic, in clinical trials, or in emerging research studies to alleviate the inflamed joint environment or to promote new cartilage tissue formation. However, these therapies remain limited in their duration and effectiveness. For this reason, current efforts are focused on improving the localization, retention, and activity of these bioactive factors. The purpose of this review is to highlight recent advances in drug delivery for the treatment of damaged or degenerated cartilage. First, we summarize material and modification techniques to improve the delivery of these factors to damaged tissue and enhance their retention and action within the joint environment. Second, we discuss recent studies using novel methods to promote new cartilage formation via biofactor delivery, that have potential for improving future long-term clinical outcomes. Lastly, we review the emerging field of orthobiologics, using delivered and endogenous cells as drug-delivering "factories" to preserve and restore joint health. Enhancing drug delivery systems can improve both restorative and regenerative treatments for damaged cartilage. STATEMENT OF SIGNIFICANCE: Articular cartilage is a remarkable and sophisticated tissue that tolerates complex stresses within the joint. When injured, cartilage cannot self-repair, and these injuries often progress to joint-wide osteoarthritis, causing patients debilitating pain and loss of mobility. Current palliative and surgical treatments only provide short-term symptomatic relief and are limited with regards to efficiency and efficacy. Bioactive factors, such as drugs and growth factors, can improve outcomes to either stabilize the degenerated environment or regenerate replacement tissue. This review highlights recent advances and novel techniques to enhance the delivery, localization, retention, and activity of these factors, providing an overview of the cartilage drug delivery field that can guide future research in restorative and regenerative treatments for damaged cartilage.
Collapse
Affiliation(s)
- Jay M Patel
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, United States
| | - Kamiel S Saleh
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, United States
| | - Jason A Burdick
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, United States; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, United States; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
35
|
Schneider MC, Chu S, Randolph MA, Bryant SJ. An in vitro and in vivo comparison of cartilage growth in chondrocyte-laden matrix metalloproteinase-sensitive poly(ethylene glycol) hydrogels with localized transforming growth factor β3. Acta Biomater 2019; 93:97-110. [PMID: 30914256 DOI: 10.1016/j.actbio.2019.03.046] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/25/2022]
Abstract
While matrix-assisted autologous chondrocyte implantation has emerged as a promising therapy to treat focal chondral defects, matrices that support regeneration of hyaline cartilage remain challenging. The goal of this work was to investigate the potential of a matrix metalloproteinase (MMP)-sensitive poly(ethylene glycol) (PEG) hydrogel containing the tethered growth factor, transforming growth factor β3 (TGF-β3), and compare cartilage regeneration in vitro and in vivo. The in vitro environment comprised chemically-defined medium while the in vivo environment utilized the subcutaneous implant model in athymic mice. Porcine chondrocytes were isolated and expanded in 2D culture for 10 days prior to encapsulation. The presence of tethered TGF-β3 reduced cell spreading. Chondrocyte-laden hydrogels were analyzed for total sulfated glycosaminoglycan and collagen contents, MMP activity, and spatial deposition of aggrecan, decorin, biglycan, and collagens type II and I. The total amount of extracellular matrix (ECM) deposited in the hydrogel constructs was similar in vitro and in vivo. However, the in vitro environment was not able to support long-term culture up to 64 days of the engineered cartilage leading to the eventual breakdown of aggrecan. The in vivo environment, on the other hand, led to more elaborate ECM, which correlated with higher MMP activity, and an overall higher quality of engineered tissue that was rich in aggrecan, decorin, biglycan and collagen type II with minimal collagen type I. Overall, the MMP-sensitive PEG hydrogel containing tethered TGF-β3 is a promising matrix for hyaline cartilage regeneration in vivo. STATEMENT OF SIGNIFICANCE: Regenerating hyaline cartilage remains a significant clinical challenge. The resultant repair tissue is often fibrocartilage, which long-term cannot be sustained. The goal of this study was to investigate the potential of a synthetic hydrogel matrix containing peptide crosslinks that can be degraded by enzymes secreted by encapsulated cartilage cells (i.e., chondrocytes) and tethered growth factors, specifically TGF-β3, to provide localized chondrogenic cues to the cells. This hydrogel led to hyaline cartilage-like tissue growth in vitro and in vivo, with minimal formation of fibrocartilage. However, the tissue formed in vitro, could not be maintained long-term. In vivo this hydrogel shows great promise as a potential matrix for use in regenerating hyaline cartilage.
Collapse
|
36
|
Browe DC, Mahon OR, Díaz‐Payno PJ, Cassidy N, Dudurych I, Dunne A, Buckley CT, Kelly DJ. Glyoxal cross‐linking of solubilized extracellular matrix to produce highly porous, elastic, and chondro‐permissive scaffolds for orthopedic tissue engineering. J Biomed Mater Res A 2019; 107:2222-2234. [DOI: 10.1002/jbm.a.36731] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Affiliation(s)
- David C. Browe
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Olwyn R. Mahon
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- School of Biochemistry and Immunology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland
| | - Pedro J. Díaz‐Payno
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Nina Cassidy
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
| | - Ivan Dudurych
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
| | - Aisling Dunne
- School of Biochemistry and Immunology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland
| | - Conor T. Buckley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| |
Collapse
|
37
|
Rathan S, Dejob L, Schipani R, Haffner B, Möbius ME, Kelly DJ. Fiber Reinforced Cartilage ECM Functionalized Bioinks for Functional Cartilage Tissue Engineering. Adv Healthc Mater 2019; 8:e1801501. [PMID: 30624015 DOI: 10.1002/adhm.201801501] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Indexed: 01/17/2023]
Abstract
Focal articular cartilage (AC) defects, if left untreated, can lead to debilitating diseases such as osteoarthritis. While several tissue engineering strategies have been developed to promote cartilage regeneration, it is still challenging to generate functional AC capable of sustaining high load-bearing environments. Here, a new class of cartilage extracellular matrix (cECM)-functionalized alginate bioink is developed for the bioprinting of cartilaginous tissues. The bioinks are 3D-printable, support mesenchymal stem cell (MSC) viability postprinting and robust chondrogenesis in vitro, with the highest levels of COLLII and ACAN expression observed in bioinks containing the highest concentration of cECM. Enhanced chondrogenesis in cECM-functionalized bioinks is also associated with progression along an endochondral-like pathway, as evident by increases in RUNX2 expression and calcium deposition in vitro. The bioinks loaded with MSCs and TGF-β3 are also found capable of supporting robust chondrogenesis, opening the possibility of using such bioinks for direct "print-and-implant" cartilage repair strategies. Finally, it is demonstrated that networks of 3D-printed polycaprolactone fibers with compressive modulus comparable to native AC can be used to mechanically reinforce these bioinks, with no loss in cell viability. It is envisioned that combinations of such biomaterials can be used in multiple-tool biofabrication strategies for the bioprinting of biomimetic cartilaginous implants.
Collapse
Affiliation(s)
- Swetha Rathan
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Léa Dejob
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Ecole Nationale Supérieure de Chimie de Mulhouse, Université de Haute-Alsace, 68200, Mulhouse, France
| | - Rossana Schipani
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
| | | | | | - Daniel J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
- Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
38
|
Zarei H, Karimpour A, Reza Khalatbary A, Talebpour Amiri F. Homing of adipose stem cells on the human amniotic membrane as a scaffold: A histological study. Int J Reprod Biomed 2019; 18:21-32. [PMID: 32043068 PMCID: PMC6996125 DOI: 10.18502/ijrm.v18i1.6193] [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: 09/05/2018] [Revised: 03/03/2019] [Accepted: 08/14/2019] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The human amniotic membrane (HAM) is a suitable and effective scaffold for cell culture and delivery, and adipose-derived stem cells (ADSCs) are an important source of stem cells for transplantation and chondrogenic differentiation. OBJECTIVE To assess the practicability of a cryopreserved HAM as a scaffold in cell proliferation and differentiation in vitro. MATERIALS AND METHODS In this experimental study, adipose tissue samples were harvested from the inguinal region of male patients aged 15-30 years. Flow cytometry was used to identify CD31, CD45, CD90, and CD105 markers in adipose stem cells. HAM was harvested from donor placenta after cesarean section, washed, trypsin-based decellularized trypsinized decellularized, and used as a scaffold via three methods: 1) ADSCs were differentiated into chondrocytes on cell culture flasks (monolayer method), and after 14 days of culture, the cells were transferred and cultured on both sides of the HAM; 2) ADSCs were cultured and differentiated directly on both sides of the HAM for 14 days (scaffold-mediated differentiation); and 3) chondrocytes were differentiated with micromass culture for 14 days, transferred on HAM, and tissue slides were histologically analyzed qualitatively. RESULTS Flow cytometry confirmed the presence of mesenchymal stem cells. Histological findings revealed that the cells adhered and grew well on the stromal layer of HAM. Among the three methods, scaffold-mediated differentiation of ADSCs showed the best results. CONCLUSION ADSCs have excellent attachment, viability, and differentiation capacity in the stromal side of HAM. Additionally, the direct culture and differentiation of ADSCs on HAM is more suitable than the culture of differentiated cells on HAM.
Collapse
Affiliation(s)
- Hooman Zarei
- Department of Anatomy, Faculty of Medicine, Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abbasali Karimpour
- Department of Anatomy, Faculty of Medicine, Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Reza Khalatbary
- Department of Anatomy, Faculty of Medicine, Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fereshteh Talebpour Amiri
- Department of Anatomy, Faculty of Medicine, Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
39
|
Nürnberger S, Schneider C, van Osch G, Keibl C, Rieder B, Monforte X, Teuschl A, Mühleder S, Holnthoner W, Schädl B, Gahleitner C, Redl H, Wolbank S. Repopulation of an auricular cartilage scaffold, AuriScaff, perforated with an enzyme combination. Acta Biomater 2019; 86:207-222. [PMID: 30590183 DOI: 10.1016/j.actbio.2018.12.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/14/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022]
Abstract
Biomaterials currently in use for articular cartilage regeneration do not mimic the composition or architecture of hyaline cartilage, leading to the formation of repair tissue with inferior characteristics. In this study we demonstrate the use of "AuriScaff", an enzymatically perforated bovine auricular cartilage scaffold, as a novel biomaterial for repopulation with regenerative cells and for the formation of high-quality hyaline cartilage. AuriScaff features a traversing channel network, generated by selective depletion of elastic fibers, enabling uniform repopulation with therapeutic cells. The complex collagen type II matrix is left intact, as observed by immunohistochemistry, SEM and TEM. The compressive modulus is diminished, but three times higher than in the clinically used collagen type I/III scaffold that served as control. Seeding tests with human articular chondrocytes (hAC) alone and in co-culture with human adipose-derived stromal/stem cells (ASC) confirmed that the network enabled cell migration throughout the scaffold. It also guides collagen alignment along the channels and, due to the generally traverse channel alignment, newly deposited cartilage matrix corresponds with the orientation of collagen within articular cartilage. In an osteochondral plug model, AuriScaff filled the complete defect with compact collagen type II matrix and enabled chondrogenic differentiation inside the channels. Using adult articular chondrocytes from bovine origin (bAC), filling of even deep defects with high-quality hyaline-like cartilage was achieved after 6 weeks in vivo. With its composition and spatial organization, AuriScaff provides an optimal chondrogenic environment for therapeutic cells to treat cartilage defects and is expected to improve long-term outcome by channel-guided repopulation followed by matrix deposition and alignment. STATEMENT OF SIGNIFICANCE: After two decades of tissue engineering for cartilage regeneration, there is still no optimal strategy available to overcome problems such as inconsistent clinical outcome, early and late graft failures. Especially large defects are dependent on biomaterials and their scaffolding, guiding and protective function. Considering the currently used biomaterials, structure and mechanical properties appear to be insufficient to fulfill this task. The novel scaffold developed within this study is the first approach enabling the use of dense cartilage matrix, repopulate it via channels and provide the cells with a compact collagen type II environment. Due to its density, it also provides better mechanical properties than materials currently used in clinics. We therefore think, that the auricular cartilage scaffold (AuriScaff) has a high potential to improve future cartilage regeneration approaches.
Collapse
|
40
|
Cunniffe GM, Díaz-Payno PJ, Sheehy EJ, Critchley SE, Almeida HV, Pitacco P, Carroll SF, Mahon OR, Dunne A, Levingstone TJ, Moran CJ, Brady RT, O'Brien FJ, Brama PA, Kelly DJ. Tissue-specific extracellular matrix scaffolds for the regeneration of spatially complex musculoskeletal tissues. Biomaterials 2019; 188:63-73. [DOI: 10.1016/j.biomaterials.2018.09.044] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/18/2018] [Accepted: 09/28/2018] [Indexed: 01/09/2023]
|
41
|
Zhang X, Zhai C, Fei H, Liu Y, Wang Z, Luo C, Zhang J, Ding Y, Xu T, Fan W. Composite Silk-Extracellular Matrix Scaffolds for Enhanced Chondrogenesis of Mesenchymal Stem Cells. Tissue Eng Part C Methods 2018; 24:645-658. [PMID: 30351193 DOI: 10.1089/ten.tec.2018.0199] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Xiao Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenjun Zhai
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Orthopedics, Yixing People's Hospital, Yixing, China
| | - Hao Fei
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhen Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chunyang Luo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiyong Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanzi Ding
- Department of Cardiovascular, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weimin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Liou JJ, Rothrauff BB, Alexander PG, Tuan RS. Effect of Platelet-Rich Plasma on Chondrogenic Differentiation of Adipose- and Bone Marrow-Derived Mesenchymal Stem Cells. Tissue Eng Part A 2018; 24:1432-1443. [DOI: 10.1089/ten.tea.2018.0065] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Jr-Jiun Liou
- Department of Bioengineering, Swanson School of Engineering, Pittsburgh, Pennsylvania
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, Pittsburgh, Pennsylvania
| | - Benjamin B. Rothrauff
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, Pittsburgh, Pennsylvania
| | - Peter G. Alexander
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, Pittsburgh, Pennsylvania
| | - Rocky S. Tuan
- Department of Bioengineering, Swanson School of Engineering, Pittsburgh, Pennsylvania
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
43
|
Rowland CR, Glass KA, Ettyreddy AR, Gloss CC, Matthews JRL, Huynh NPT, Guilak F. Regulation of decellularized tissue remodeling via scaffold-mediated lentiviral delivery in anatomically-shaped osteochondral constructs. Biomaterials 2018; 177:161-175. [PMID: 29894913 PMCID: PMC6082159 DOI: 10.1016/j.biomaterials.2018.04.049] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 01/25/2023]
Abstract
Cartilage-derived matrix (CDM) has emerged as a promising scaffold material for tissue engineering of cartilage and bone due to its native chondroinductive capacity and its ability to support endochondral ossification. Because it consists of native tissue, CDM can undergo cellular remodeling, which can promote integration with host tissue and enables it to be degraded and replaced by neotissue over time. However, enzymatic degradation of decellularized tissues can occur unpredictably and may not allow sufficient time for mechanically competent tissue to form, especially in the harsh inflammatory environment of a diseased joint. The goal of the current study was to engineer cartilage and bone constructs with the ability to inhibit aberrant inflammatory processes caused by the cytokine interleukin-1 (IL-1), through scaffold-mediated delivery of lentiviral particles containing a doxycycline-inducible IL-1 receptor antagonist (IL-1Ra) transgene on anatomically-shaped CDM constructs. Additionally, scaffold-mediated lentiviral gene delivery was used to facilitate spatial organization of simultaneous chondrogenic and osteogenic differentiation via site-specific transduction of a single mesenchymal stem cell (MSC) population to overexpress either chondrogenic, transforming growth factor-beta 3 (TGF-β3), or osteogenic, bone morphogenetic protein-2 (BMP-2), transgenes. Controlled induction of IL-1Ra expression protected CDM hemispheres from inflammation-mediated degradation, and supported robust bone and cartilage tissue formation even in the presence of IL-1. In the absence of inflammatory stimuli, controlled cellular remodeling was exploited as a mechanism for fusing concentric CDM hemispheres overexpressing BMP-2 and TGF-β3 into a single bi-layered osteochondral construct. Our findings demonstrate that site-specific delivery of inducible and tunable transgenes confers spatial and temporal control over both CDM scaffold remodeling and neotissue composition. Furthermore, these constructs provide a microphysiological in vitro joint organoid model with site-specific, tunable, and inducible protein delivery systems for examining the spatiotemporal response to pro-anabolic and/or inflammatory signaling across the osteochondral interface.
Collapse
Affiliation(s)
- Christopher R Rowland
- Washington University in Saint Louis, Saint Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA
| | | | | | - Catherine C Gloss
- Washington University in Saint Louis, Saint Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA
| | - Jared R L Matthews
- Washington University in Saint Louis, Saint Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA
| | - Nguyen P T Huynh
- Washington University in Saint Louis, Saint Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Duke University, Durham, NC 27710, USA
| | - Farshid Guilak
- Washington University in Saint Louis, Saint Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
44
|
Controlled Non-Viral Gene Delivery in Cartilage and Bone Repair: Current Strategies and Future Directions. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
45
|
Sasaki H, Rothrauff BB, Alexander PG, Lin H, Gottardi R, Fu FH, Tuan RS. In Vitro Repair of Meniscal Radial Tear With Hydrogels Seeded With Adipose Stem Cells and TGF-β3. Am J Sports Med 2018; 46:2402-2413. [PMID: 30001494 DOI: 10.1177/0363546518782973] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Radial tears of the meniscus are a common knee injury, frequently resulting in osteoarthritis. To date, there are no established, effective treatments for radial tears. Adipose-derived stem cells (ASCs) may be an attractive cell source for meniscal regeneration because they can be quickly isolated in large number and are capable of undergoing induced fibrochondrogenic differentiation mediated by transforming growth factor β3 (TGF-β3). However, the use of ASCs for meniscal repair is largely unexplored. HYPOTHESIS ASC-seeded hydrogels with preloaded TGF-β3 will improve meniscal healing of radial tears, as modeled in an explant model. STUDY DESIGN Controlled laboratory study. METHODS With an institutional review board-exempted protocol, human ASCs were isolated from the infrapatellar fat pads of 3 donors, obtained after total knee replacement, and characterized. ASCs were encapsulated in photocrosslinkable methacrylated gelatin hydrogels to form 3-dimensional constructs, which were placed into tissue culture. The effect of TGF-β3-whether preloaded into the hydrogel or added as a soluble medium supplement-on matrix-sulfated proteoglycan deposition in the constructs was evaluated. A meniscal explant culture model was used to simulate meniscal repair. Cylindrical-shaped explants were excised from the inner avascular region of adult bovine menisci, and a radial tear was modeled by cutting perpendicular to the meniscal main fibers to the length of the radius. Six combinations of hydrogels-namely, acellular and ASC-seeded hydrogels supplemented with preloaded TGF-β3 (2 µg/mL) or soluble TGF-β3 (10 ng/mL) and without supplement-were injected into the radial tear and stabilized by photocrosslinking with visible light. At 4 and 8 weeks of culture, healing was assessed through histology, immunofluorescence staining, and mechanical testing. RESULTS ASCs isolated from the 3 donors exhibited colony-forming and multilineage differentiation potential. Hydrogels preloaded with TGF-β3 and those cultured in soluble TGF-β3 showed robust matrix-sulfated proteoglycan deposition. ASC-seeded hydrogels promoted superior healing as compared with acellular hydrogels, with preloaded or soluble TGF-β3 further improving histological scores and mechanical properties. CONCLUSION These findings demonstrated that ASC-seeded hydrogels preloaded with TGF-β3 enhanced healing of radial meniscal tears in an in vitro meniscal repair model. CLINICAL RELEVANCE Injection delivery of ASCs in a TGF-β3-preloaded photocrosslinkable hydrogel represents a novel candidate strategy to repair meniscal radial tears and minimize further osteoarthritic joint degeneration.
Collapse
Affiliation(s)
| | | | | | - Hang Lin
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Freddie H Fu
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rocky S Tuan
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
46
|
Sun Y, Yan L, Chen S, Pei M. Functionality of decellularized matrix in cartilage regeneration: A comparison of tissue versus cell sources. Acta Biomater 2018; 74:56-73. [PMID: 29702288 PMCID: PMC7307012 DOI: 10.1016/j.actbio.2018.04.048] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 01/12/2023]
Abstract
Increasing evidence indicates that decellularized extracellular matrices (dECMs) derived from cartilage tissues (T-dECMs) or chondrocytes/stem cells (C-dECMs) can support proliferation and chondrogenic differentiation of cartilage-forming cells. However, few review papers compare the differences between these dECMs when they serve as substrates for cartilage regeneration. In this review, after an introduction of cartilage immunogenicity and decellularization methods to prepare T-dECMs and C-dECMs, a comprehensive comparison focuses on the effects of T-dECMs and C-dECMs on proliferation and chondrogenic differentiation of chondrocytes/stem cells in vitro and in vivo. Key factors within dECMs, consisting of microarchitecture characteristics and micromechanical properties as well as retained insoluble and soluble matrix components, are discussed in-depth for potential mechanisms underlying the functionality of these dECMs in regulating chondrogenesis. With this information, we hope to benefit dECM based cartilage engineering and tissue regeneration for future clinical application. STATEMENT OF SIGNIFICANCE The use of decellularized extracellular matrix (dECM) is becoming a promising approach for tissue engineering and regeneration. Compared to dECM derived from cartilage tissue, recently reported dECM from cell sources exhibits a distinct role in cell based cartilage regeneration. In this review paper, for the first time, tissue and cell based dECMs are comprehensively compared for their functionality in cartilage regeneration. This information is expected to provide an update for dECM based cartilage regeneration.
Collapse
Affiliation(s)
- Yu Sun
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Lianqi Yan
- Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan 610083, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
47
|
Sivandzade F, Mashayekhan S. Design and fabrication of injectable microcarriers composed of acellular cartilage matrix and chitosan. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2018; 29:683-700. [DOI: 10.1080/09205063.2018.1433422] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Farzane Sivandzade
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
48
|
Dale TP, Forsyth NR. Ectopic Telomerase Expression Fails to Maintain Chondrogenic Capacity in Three-Dimensional Cultures of Clinically Relevant Cell Types. Biores Open Access 2018; 7:10-24. [PMID: 29588876 PMCID: PMC5865620 DOI: 10.1089/biores.2018.0008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The poor healing capacity of cartilage and lack of effective treatment for associated disease and trauma makes it a strong candidate for a regenerative medicine approach. Potential therapies tested to date, although effective, have met with a number of intrinsic difficulties possibly related to limited autologous chondrocyte cell yield and quality of cartilage produced. A potential mechanism to bypass limited cell yields and improve quality of differentiation is to immortalize relevant cell types through the ectopic expression of telomerase. Pellet cultures of human chondrocytes (OK3), bone marrow mesenchymal stem cells (BMA13), and embryonic stem cell (H1 line)-derived cells (1C6) and their human telomerase reverse transcriptase (hTERT) transduced counterparts were maintained for 20 days in standard maintenance medium (MM) or transforming growth factor-β3-supplemented prochondrogenic medium (PChM). Pellets were assessed for volume and density by microcomputed tomography. Quantitative gene expression (COL1A2, COL2A1, COL3A1, COL6A3, COL10A1, ACAN, COMP, SOX9); sulfated glycosaminoglycans (sGAGs), and DNA quantification were performed. Histology and immunohistochemistry were used to determine matrix constituent distribution. Pellet culture in PChM resulted in significantly larger pellets with an overall increased density when compared with MM culture. Gene expression analysis revealed similarities in expression patterns between telomerase-transduced and parental cells in both MM and PChM. Of the three parental cell types examined OK3 and BMA13 produced similar amounts of pellet-associated sGAG in PChM (4.62 ± 1.20 and 4.91 ± 1.37 μg, respectively) with lower amounts in 1C6 (2.89 ± 0.52 μg), corresponding to 3.1, 2.3, and 1.6-fold increases from day 0. In comparison, telomerase-transduced cells all had much lower sGAG with OK3H at 2.74 ± 0.11 μg, BMA13H 1.29 ± 0.34 μg, and 1C6H 0.52 ± 0.01 μg corresponding to 1.2, 0.87, and 0.34-fold changes compared with day 0. Histology of day 20 pellets displayed reduced staining overall for collagens and sGAG in telomerase-transduced cells, most notably with alterations in aggrecan and collagen VI; all cells stained positively for collagen II. We conclude that while telomerase transduction may be an effective technique to extend cellular proliferative capacity, it is not sufficient in isolation to sustain a naive chondrogenic phenotype across multiple cell types.
Collapse
Affiliation(s)
- Tina P Dale
- Faculty of Medicine and Health Sciences, Guy Hilton Research Center, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| | - Nicholas R Forsyth
- Faculty of Medicine and Health Sciences, Guy Hilton Research Center, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| |
Collapse
|
49
|
Daly AC, Sathy BN, Kelly DJ. Engineering large cartilage tissues using dynamic bioreactor culture at defined oxygen conditions. J Tissue Eng 2018; 9:2041731417753718. [PMID: 29399319 PMCID: PMC5788092 DOI: 10.1177/2041731417753718] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/22/2017] [Indexed: 11/23/2022] Open
Abstract
Mesenchymal stem cells maintained in appropriate culture conditions are capable of producing robust cartilage tissue. However, gradients in nutrient availability that arise during three-dimensional culture can result in the development of spatially inhomogeneous cartilage tissues with core regions devoid of matrix. Previous attempts at developing dynamic culture systems to overcome these limitations have reported suppression of mesenchymal stem cell chondrogenesis compared to static conditions. We hypothesize that by modulating oxygen availability during bioreactor culture, it is possible to engineer cartilage tissues of scale. The objective of this study was to determine whether dynamic bioreactor culture, at defined oxygen conditions, could facilitate the development of large, spatially homogeneous cartilage tissues using mesenchymal stem cell laden hydrogels. A dynamic culture regime was directly compared to static conditions for its capacity to support chondrogenesis of mesenchymal stem cells in both small and large alginate hydrogels. The influence of external oxygen tension on the response to the dynamic culture conditions was explored by performing the experiment at 20% O2 and 3% O2. At 20% O2, dynamic culture significantly suppressed chondrogenesis in engineered tissues of all sizes. In contrast, at 3% O2 dynamic culture significantly enhanced the distribution and amount of cartilage matrix components (sulphated glycosaminoglycan and collagen II) in larger constructs compared to static conditions. Taken together, these results demonstrate that dynamic culture regimes that provide adequate nutrient availability and a low oxygen environment can be employed to engineer large homogeneous cartilage tissues. Such culture systems could facilitate the scaling up of cartilage tissue engineering strategies towards clinically relevant dimensions.
Collapse
Affiliation(s)
- Andrew C Daly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Binulal N Sathy
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Daniel J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
50
|
Armiento AR, Stoddart MJ, Alini M, Eglin D. Biomaterials for articular cartilage tissue engineering: Learning from biology. Acta Biomater 2018; 65:1-20. [PMID: 29128537 DOI: 10.1016/j.actbio.2017.11.021] [Citation(s) in RCA: 363] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/05/2017] [Accepted: 11/07/2017] [Indexed: 12/27/2022]
Abstract
Articular cartilage is commonly described as a tissue that is made of up to 80% water, is devoid of blood vessels, nerves, and lymphatics, and is populated by only one cell type, the chondrocyte. At first glance, an easy tissue for clinicians to repair and for scientists to reproduce in a laboratory. Yet, chondral and osteochondral defects currently remain an open challenge in orthopedics and tissue engineering of the musculoskeletal system, without considering osteoarthritis. Why do we fail in repairing and regenerating articular cartilage? Behind its simple and homogenous appearance, articular cartilage hides a heterogeneous composition, a high level of organisation and specific biomechanical properties that, taken together, make articular cartilage a unique material that we are not yet able to repair or reproduce with high fidelity. This review highlights the available therapies for cartilage repair and retraces the research on different biomaterials developed for tissue engineering strategies. Their potential to recreate the structure, including composition and organisation, as well as the function of articular cartilage, intended as cell microenvironment and mechanically competent replacement, is described. A perspective of the limitations of the current research is given in the light of the emerging technologies supporting tissue engineering of articular cartilage. STATEMENT OF SIGNIFICANCE The mechanical properties of articular tissue reflect its functionally organised composition and the recreation of its structure challenges the success of in vitro and in vivo reproduction of the native cartilage. Tissue engineering and biomaterials science have revolutionised the way scientists approach the challenge of articular cartilage repair and regeneration by introducing the concept of the interdisciplinary approach. The clinical translation of the current approaches are not yet fully successful, but promising results are expected from the emerging and developing new generation technologies.
Collapse
Affiliation(s)
- A R Armiento
- AO Research Institute Davos, Davos Platz, Switzerland.
| | - M J Stoddart
- AO Research Institute Davos, Davos Platz, Switzerland; University Medical Center, Albert-Ludwigs University, Freiburg, Germany.
| | - M Alini
- AO Research Institute Davos, Davos Platz, Switzerland.
| | - D Eglin
- AO Research Institute Davos, Davos Platz, Switzerland.
| |
Collapse
|