1
|
van Mourik M, Tiemeijer BM, van Zon M, Abinzano F, Tel J, Foolen J, Ito K. Cartilage-derived cells display heterogeneous pericellular matrix synthesis in agarose microgels. Matrix Biol Plus 2024; 23:100157. [PMID: 39139760 PMCID: PMC11321428 DOI: 10.1016/j.mbplus.2024.100157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 08/15/2024] Open
Abstract
The pericellular matrix (PCM) surrounding chondrocytes is essential for articular cartilage tissue engineering. As the current isolation methods to obtain chondrocytes with their PCM (chondrons) result in a heterogeneous mixture of chondrocytes and chondrons, regenerating the PCM using a tissue engineering approach could prove beneficial. In this study, we aimed to discern the behavior of articular chondrocytes (ACs) in regenerating the PCM in such an approach and whether this would also be true for articular cartilage-derived progenitor cells (ACPCs), as an alternative cell source. Bovine ACs and ACPCs were encapsulated in agarose microgels using droplet-based microfluidics. ACs were stimulated with TGF-β1 and dexamethasone and ACPCs were sequentially stimulated with BMP-9 followed by TGF-β1 and dexamethasone. After 0, 3, 5, and 10 days of culture, PCM components, type-VI collagen and perlecan, and ECM component, type-II collagen, were assessed using flow cytometry and fluorescence microscopy. Both ACs and ACPCs synthesized the PCM before the ECM. It was seen for the first time that synthesis of type-VI collagen always preceded perlecan. While the PCM synthesized by ACs resembled native chondrons after only 5 days of culture, ACPCs often made less well-structured PCMs. Both cell types showed variations between individual cells and donors. On one hand, this was more prominent in ACPCs, but also a subset of ACPCs showed superior PCM and ECM regeneration, suggesting that isolating these cells may potentially improve cartilage repair strategies.
Collapse
Affiliation(s)
- Marloes van Mourik
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Bart M. Tiemeijer
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Maarten van Zon
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
| | - Florencia Abinzano
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Jurjen Tel
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Jasper Foolen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| |
Collapse
|
2
|
Petta D, D'Arrigo D, Salehi S, Talò G, Bonetti L, Vanoni M, Deabate L, De Nardo L, Dubini G, Candrian C, Moretti M, Lopa S, Arrigoni C. A personalized osteoarthritic joint-on-a-chip as a screening platform for biological treatments. Mater Today Bio 2024; 26:101072. [PMID: 38757057 PMCID: PMC11097088 DOI: 10.1016/j.mtbio.2024.101072] [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: 11/29/2023] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Osteoarthritis (OA) is a highly disabling pathology, characterized by synovial inflammation and cartilage degeneration. Orthobiologics have shown promising results in OA treatment thanks to their ability to influence articular cells and modulate the inflammatory OA environment. Considering their complex mechanism of action, the development of reliable and relevant joint models appears as crucial to select the best orthobiologics for each patient. The aim of this study was to establish a microfluidic OA model to test therapies in a personalized human setting. The joint-on-a-chip model included cartilage and synovial compartments, containing hydrogel-embedded chondrocytes and synovial fibroblasts, separated by a channel for synovial fluid. For the cartilage compartment, a Hyaluronic Acid-based matrix was selected to preserve chondrocyte phenotype. Adding OA synovial fluid induced the production of inflammatory cytokines and degradative enzymes, generating an OA microenvironment. Personalized models were generated using patient-matched cells and synovial fluid to test the efficacy of mesenchymal stem cells on OA signatures. The patient-specific models allowed monitoring changes induced by cell injection, highlighting different individual responses to the treatment. Altogether, these results support the use of this joint-on-a-chip model as a prognostic tool to screen the patient-specific efficacy of orthobiologics.
Collapse
Affiliation(s)
- Dalila Petta
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
| | - Daniele D'Arrigo
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- ISBE-SYSBIO Centre of Systems Biology, Milan, Italy at Department of Biotechnology and Biosciences, Università Degli Studi di Milano Bicocca, Piazza Della Scienza 2, 20126, Milan, Italy
| | - Shima Salehi
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
| | - Giuseppe Talò
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
| | - Lorenzo Bonetti
- Department of Chemistry, Materials and Chemical Engineering G.Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Marco Vanoni
- ISBE-SYSBIO Centre of Systems Biology, Milan, Italy at Department of Biotechnology and Biosciences, Università Degli Studi di Milano Bicocca, Piazza Della Scienza 2, 20126, Milan, Italy
| | - Luca Deabate
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
| | - Luigi De Nardo
- Department of Chemistry, Materials and Chemical Engineering G.Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Gabriele Dubini
- Department of Chemistry, Materials and Chemical Engineering G.Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Christian Candrian
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana (USI), Via Buffi 13, 6900, Lugano, Switzerland
| | - Matteo Moretti
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
- Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana (USI), Via Buffi 13, 6900, Lugano, Switzerland
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
| | - Chiara Arrigoni
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana (USI), Via Buffi 13, 6900, Lugano, Switzerland
| |
Collapse
|
3
|
Lee MFH, Steffens D, Chung JHY, Posniak S, Cheng K, Clark J, Wallace G, Mukherjee P. Coculture of Chondrocytes and Stem Cells: A Review of Head and Neck Cell Lines for Cartilage Regeneration. Cells Tissues Organs 2024; 214:36-51. [PMID: 38513621 DOI: 10.1159/000538461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 03/16/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION Bioprinting, using "bio-inks" consisting of living cells, supporting structures, and biological motifs to create customized constructs, is an emerging technique that aims to overcome the challenges of cartilaginous reconstruction of head and neck structures. Several living cell lines and culturing methods have been explored as bio-inks with varying efficacy. Coculture of primary chondrocytes and stem cells (SCs) is one technique well established for degenerative joint disease treatment, with potential for use in expanding chondrocyte populations for bio-inks. This study aimed to evaluate the techniques for coculture of primary chondrocytes and SCs for head and neck cartilage regeneration. METHODS A literature review was performed through OVID/Web of Science/MEDLINE/BIOSIS Previews/Embase. Studies reporting on chondrocytes and SCs in conjunction with coculture or cartilage regeneration were included. Studies not reporting on findings from chondrocytes/SCs of the head and neck were excluded. Extracted data included cell sources, coculture ratios, and histological, biochemical, and clinical outcomes. RESULTS Fifteen studies met inclusion criteria. Auricular cartilage was the most common chondrocyte source (n = 10), then nasal septum (n = 5), articular (n = 1), and tracheal cartilage (n = 1). Bone marrow was the most common SC source (n = 9) then adipose tissue (n = 7). Techniques varied, with coculture ratios ranging from 1:1 to 1:10. All studies reported coculture to be superior to SC monoculture by all outcomes. Most studies reported superiority or equivalence of coculture to chondrocyte monoculture by all outcomes. When comparing clinical outcomes, coculture constructs were equivalent to chondrocyte monoculture in diameter and equivalent or inferior in wet weight and height. CONCLUSION Coculture of primary chondrocytes and SCs is a promising technique for expanding chondrocyte populations, with at least equivalence to chondrocyte monoculture and superior to SC monoculture when seeded at the same chondrocyte densities. However, there remains a lack of consensus regarding the optimal cell sources and coculture ratios.
Collapse
Affiliation(s)
- Michael Fook-Ho Lee
- Royal Prince Alfred Institute of Academic Surgery (RPA-IAS), Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Daniel Steffens
- Royal Prince Alfred Institute of Academic Surgery (RPA-IAS), Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
- Surgical Outcomes Research Centre (SOuRCe), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Johnson H Y Chung
- Intelligent Polymer Research Institute, University of Wollongong, Wollongong, New South Wales, Australia
| | - Steven Posniak
- Intelligent Polymer Research Institute, University of Wollongong, Wollongong, New South Wales, Australia
| | - Kai Cheng
- Royal Prince Alfred Institute of Academic Surgery (RPA-IAS), Sydney, New South Wales, Australia
| | - Jonathan Clark
- Royal Prince Alfred Institute of Academic Surgery (RPA-IAS), Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Gordon Wallace
- Intelligent Polymer Research Institute, University of Wollongong, Wollongong, New South Wales, Australia
| | - Payal Mukherjee
- Royal Prince Alfred Institute of Academic Surgery (RPA-IAS), Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
- Department of Medicine, Health and Human Sciences, Department of Clinical Medicine, Macquarie University, Macquarie Park, New South Wales, Australia
| |
Collapse
|
4
|
Banihashemian SA, Zamanlui Benisi S, Hosseinzadeh S, Shojaei S, Abbaszadeh HA. Chitosan/Hyaluronan and Alginate-Nanohydroxyapatite Biphasic Scaffold as a Promising Matrix for Osteoarthritis Disorders. Adv Pharm Bull 2024; 14:176-191. [PMID: 38585453 PMCID: PMC10997938 DOI: 10.34172/apb.2024.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/24/2023] [Accepted: 07/19/2023] [Indexed: 04/09/2024] Open
Abstract
Purpose Regenerative medicine offers new techniques for osteoarthritis (OA) disorders, especially while considering simultaneous chondral and subchondral regenerations. Methods Chitosan and hyaluronan were chemically bound as the chondral phase and the osteogenic layer was prepared with alginate and nano-hydroxyapatite (nHAP). These scaffolds were fixed by fibrin glue as a biphasic scaffold and then examined. Results Scanning electron microscopy (SEM) confirmed the porosity of 61.45±4.51 and 44.145±2.81 % for the subchondral and chondral layers, respectively. The composition analysis by energy dispersive X-ray (EDAX) indicated the various elements of both hydrogels. Also, their mechanical properties indicated that the highest modulus and resistance values corresponded to the biphasic hydrogel as 108.33±5.56 and 721.135±8.21 kPa, despite the same strain value as other groups. Their individual examinations demonstrated the proteoglycan synthesis of the chondral layer and also, the alkaline phosphatase (ALP) activity of the subchondral layer as 13.3±2.2 ng. After 21 days, the cells showed a mineralized surface and a polygonal phenotype, confirming their commitment to bone and cartilage tissues, respectively. Immunostaining of collagen I and II represented greater extracellular matrix (ECM) secretion in the biphasic composite group due to the paracrine effect of the two cell types on each other. Conclusion For the first time, the ability of this biphasic scaffold to regenerate both tissue types was evaluated and the results showed satisfactory cellular commitment to bone and cartilage tissues. Thus, this scaffold can be considered a new strategy for the preparation of implants for OA.
Collapse
Affiliation(s)
- Seyed Abdolvahab Banihashemian
- Advanced Medical Sciences and Technologies Department, Faculty of Biomedical Engineering, Central Tehran Branch Islamic Azad University, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Tehran Central Branch, Islamic Azad University, Tehran, Iran
| | - Simzar Hosseinzadeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahrokh Shojaei
- Islamic Azad University Central Tehran Branch, Department of Biomedical Engineering, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Asadian M, Tomasina C, Onyshchenko Y, Chan KV, Norouzi M, Zonderland J, Camarero-Espinosa S, Morent R, De Geyter N, Moroni L. The role of plasma-induced surface chemistry on polycaprolactone nanofibers to direct chondrogenic differentiation of human mesenchymal stem cells. J Biomed Mater Res A 2024; 112:210-230. [PMID: 37706337 DOI: 10.1002/jbm.a.37607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023]
Abstract
Bone marrow-derived mesenchymal stromal cells (BMSCs) are extensively being utilized for cartilage regeneration owing to their excellent differentiation potential and availability. However, controlled differentiation of BMSCs towards cartilaginous phenotypes to heal full-thickness cartilage defects remains challenging. This study investigates how different surface properties induced by either coating deposition or biomolecules immobilization onto nanofibers (NFs) could affect BMSCs chondro-inductive behavior. Accordingly, electrospun poly(ε-caprolactone) (PCL) NFs were exposed to two surface modification strategies based on medium-pressure plasma technology. The first strategy is plasma polymerization, in which cyclopropylamine (CPA) or acrylic acid (AcAc) monomers were plasma polymerized to obtain amine- or carboxylic acid-rich NFs, respectively. The second strategy uses a combination of CPA plasma polymerization and a post-chemical technique to immobilize chondroitin sulfate (CS) onto the NFs. These modifications could affect surface roughness, hydrophilicity, and chemical composition while preserving the NFs' nano-morphology. The results of long-term BMSCs culture in both basic and chondrogenic media proved that the surface modifications modulated BMSCs chondrogenic differentiation. Indeed, the incorporation of polar groups by different modification strategies had a positive impact on the cell proliferation rate, production of the glycosaminoglycan matrix, and expression of extracellular matrix proteins (collagen I and collagen II). The chondro-inductive behavior of the samples was highly dependent on the nature of the introduced polar functional groups. Among all samples, carboxylic acid-rich NFs promoted chondrogenesis by higher expression of aggrecan, Sox9, and collagen II with downregulation of hypertrophic markers. Hence, this approach showed an intrinsic potential to have a non-hypertrophic chondrogenic cell phenotype.
Collapse
Affiliation(s)
- Mahtab Asadian
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
- Prometheus Division of Skeletal Tissue Engineering, Department of Materials Science, KU Leuven University, Leuven, Belgium
| | - Clarissa Tomasina
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
| | - Yuliia Onyshchenko
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Ke Vin Chan
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Mohammad Norouzi
- Department of Pharmacology, University of Montreal, Montreal, Québec, Canada
| | - Jip Zonderland
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
| | - Sandra Camarero-Espinosa
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
- POLYMAT University of the Basque Country UPV/EHU Avenida Tolosa 72, Donostia/San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science, Euskadi Pl. 5, Bilbao, Spain
| | - Rino Morent
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Nathalie De Geyter
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
6
|
Esmaeili A, Hosseini S, Baghaban Eslaminejad M. Co-culture engineering: a promising strategy for production of engineered extracellular vesicle for osteoarthritis treatment. Cell Commun Signal 2024; 22:29. [PMID: 38200606 PMCID: PMC10782541 DOI: 10.1186/s12964-023-01467-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
The therapeutic effects of extracellular vesicles (EVs) have been identified as a significant factor in intercellular communication in different disease treatments, including osteoarthritis (OA). Compared to the conventional approaches in treating OA, EV therapy is a non-invasive and cell-free method. However, improving the yield of EVs and their therapeutic effects are the main challenges for clinical applications. In this regard, researchers are using the EV engineering potential to overcome these challenges. New findings suggest that the co-culture strategy as an indirect EV engineering method efficiently increases EV production and quality. The co-culture of mesenchymal stem cells (MSCs) and chondrocytes has improved their chondrogenesis, anti-inflammatory effects, and regenerative properties which are mediated by EVs. Hence, co-culture engineering by considerable systems could be useful in producing engineered EVs for different therapeutic applications. Here, we review various co-culture approaches, including diverse direct and indirect, 2D and 3D cell cultures, as well as static and dynamic systems. Meanwhile, we suggest and discuss the advantages of combined strategies to achieve engineered EVs for OA treatment.
Collapse
Affiliation(s)
- Abazar Esmaeili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
7
|
Burdis R, Gallostra XB, Kelly DJ. Temporal Enzymatic Treatment to Enhance the Remodeling of Multiple Cartilage Microtissues into a Structurally Organized Tissue. Adv Healthc Mater 2024; 13:e2300174. [PMID: 37858935 PMCID: PMC11468768 DOI: 10.1002/adhm.202300174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 08/29/2023] [Indexed: 10/21/2023]
Abstract
Scaffold-free tissue engineering aims to recapitulate key aspects of normal developmental processes to generate biomimetic grafts. Although functional cartilaginous tissues are engineered using such approaches, considerable challenges remain. Herein, the benefits of engineering cartilage via the fusion of multiple cartilage microtissues compared to using (millions of) individual cells to generate a cartilaginous graft are demonstrated. Key advantages include the generation of a richer extracellular matrix, more hyaline-like cartilage phenotype, and superior shape fidelity. A major drawback of aggregate engineering is that individual microtissues do not completely (re)model and remnants of their initial architectures remain throughout the macrotissue. To address this, a temporal enzymatic (chondroitinase-ABC) treatment is implemented to accelerate structural (re)modeling and shown to support robust fusion between adjacent microtissues, enhance microtissue (re)modeling, and enable the development of a more biomimetic tissue with a zonally organized collagen network. Additionally, enzymatic treatment is shown to modulate matrix composition, tissue phenotype, and to a lesser extent, tissue mechanics. This work demonstrates that microtissue self-organization is an effective method for engineering scaled-up cartilage grafts with a predefined geometry and near-native levels of matrix accumulation. Importantly, key limitations associated with using biological building blocks can be alleviated by temporal enzymatic treatment during graft development.
Collapse
Affiliation(s)
- Ross Burdis
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College DublinDublinD02 PN40Ireland
- Department of MechanicalManufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 PN40Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 PN40Ireland
| | - Xavier Barceló Gallostra
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College DublinDublinD02 PN40Ireland
- Department of MechanicalManufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 PN40Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 PN40Ireland
| | - Daniel J. Kelly
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College DublinDublinD02 PN40Ireland
- Department of MechanicalManufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 PN40Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 PN40Ireland
- Department of Anatomy and Regenerative MedicineRoyal College of Surgeons in IrelandDublinD02 YN77Ireland
| |
Collapse
|
8
|
Lp MRL, Agrawal DK. Biomechanical Forces in the Tissue Engineering and Regeneration of Shoulder, Hip, Knee, and Ankle Joints. JOURNAL OF BIOTECHNOLOGY AND BIOMEDICINE 2023; 6:491-500. [PMID: 38037618 PMCID: PMC10688570 DOI: 10.26502/jbb.2642-91280111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Tear on the tendon, ligament and articular cartilage of the joints do not heal by itself and new modalities of treatment are required to address the need for full restoration of joint functions. Accompanied by degenerative diseases, the healing of these tissues does not occur naturally and hence requires surgical interventions, but with associated morbidity. Tissue engineering strategies are now focusing on the effective incorporation of biomechanical stimulation by the application of biomechanical forces relevant to the tissue of interest to regenerate and engineer functional tissues. Bioreactors are being continuously developed to accomplish this goal. Although bioreactors have been developed, the advancement in the field of biomaterial, basic science, and cell engineering warrant further refinement for their effective use. In this article we reviewed the application of biomechanical forces in the tissue engineering and regeneration of the joints such as rotator cuff of shoulder, ball and socket joint of the hip, articular cartilage of knee, and the ankle joints.
Collapse
Affiliation(s)
- Merlin Rajesh Lal Lp
- Department of Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| | - Devendra K Agrawal
- Department of Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| |
Collapse
|
9
|
Hosseinzadeh M, Kamali A, Baghaban Eslaminejad M, Hosseini S. Higher ratios of chondrocyte to mesenchymal stem cells elevate the therapeutic effects of extracellular vesicles harvested from chondrocyte/mesenchymal stem cell co-culture on osteoarthritis in a rat model. Cell Tissue Res 2023; 394:145-162. [PMID: 37526734 DOI: 10.1007/s00441-023-03819-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023]
Abstract
Extracellular vesicles (EVs) may have a key therapeutic role and offer an innovative treatment for osteoarthritis (OA). Studies have shown that ratio of MSC/chondrocyte could affect their therapeutic outcomes. Here, we investigate the chondrogenic potential and therapeutic effect of EVs derived from MSCs and chondrocytes in the naïve, chondrogenically primed, and co-culture states to treat OA. EVs are isolated from naïve MSCs (M-EV), chondrogenically primed MSCs (cpM-EV), chondrocytes (C-EV), and co-cultures of chondrocytes plus MSCs at ratios of 1:1 (C/M-EV), 2:1 (2C/M-EV), and 4:1 (4C/M-EV). We characterized the isolated EVs in terms of surface markers, morphology, size, and zeta potential, and evaluated their chondrogenic potential in vitro by qRT-PCR and histological analyses. Next, these EVs were intra-articularly injected into osteoarthritic cartilage of a rat model and assessed by radiography, gait parameters, and histological and immunohistochemical analyses. EVs obtained from chondrocytes co-cultured with MSCs resulted in improved matrix production and functional differentiation. Our research showed that close proximity between the two cell types was essential for this response, and improved chondrogenesis and matrix formation were the outcomes of this interaction in vitro. Furthermore, in the in vivo rat OA model induced by a monoiodoacetate (MIA), we observed recovery from OA by increasing ratio of the C/M-derived EV group compared to the other groups. Our findings show that the increasing chondrocyte ratio to MSC leads to high chondrogenic induction and the therapeutic effect of harvested EVs for cartilage repair.
Collapse
Affiliation(s)
- Maryam Hosseinzadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Kamali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
10
|
Liu Y, Wu W, Seunggi C, Li Z, Huang Y, Zhou K, Wang B, Chen Z, Zhang Z. The application and progress of stem cells in auricular cartilage regeneration: a systematic review. Front Cell Dev Biol 2023; 11:1204050. [PMID: 37564374 PMCID: PMC10409996 DOI: 10.3389/fcell.2023.1204050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Background: The treatment of microtia or acquired ear deformities by surgery is a significant challenge for plastic and ENT surgeons; one of the most difficult points is constructing the scaffold for auricular reconstruction. As a type of cell with multiple differentiation potentials, stem cells play an essential role in the construction of cartilage scaffolds, and therefore have received widespread attention in ear reconstructive research. Methods: A literature search was conducted for peer-reviewed articles between 2005 and 2023 with the following keywords: stem cells; auricular cartilage; ear cartilage; conchal cartilage; auricular reconstruction, regeneration, and reparation of chondrocytes; tissue engineering in the following databases: PubMed, MEDLINE, Cochrane, and Ovid. Results: Thirty-three research articles were finally selected and their main characteristics were summarized. Adipose-derived stem cells (ADSCs), bone marrow mesenchymal stem cells (BMMSCs), perichondrial stem/progenitor cells (PPCs), and cartilage stem/progenitor cells (CSPCs) were mainly used in chondrocyte regeneration. Injecting the stem cells into the cartilage niche directly, co-culturing the stem cells with the auricular cartilage cells, and inducing the cells in the chondrogenic medium in vitro were the main methods that have been demonstrated in the studies. The chondrogenic ability of these cells was observed in vitro, and they also maintained good elasticity and morphology after implantation in vivo for a period of time. Conclusion: ADSC, BMMSC, PPC, and CSPC were the main stem cells that have been researched in craniofacial cartilage reconstruction, the regenerative cartilage performed highly similar to normal cartilage, and the test of AGA and type II collagen content also proved the cartilage property of the neo-cartilage. However, stem cell reconstruction of the auricle is still in the initial stage of animal experiments, transplantation with such scaffolds in large animals is still lacking, and there is still a long way to go.
Collapse
Affiliation(s)
- Yu Liu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Wenqing Wu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chun Seunggi
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Yeqian Huang
- West China Hospital, Sichuan University, Chengdu, China
| | - Kai Zhou
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Baoyun Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhixing Chen
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhenyu Zhang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Hunziker EB, Shintani N, Lippuner K, Vögelin E, Keel MJB. In major joint diseases the human synovium retains its potential to form repair cartilage. Sci Rep 2023; 13:10375. [PMID: 37365169 DOI: 10.1038/s41598-023-34841-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
The inner surface layer of human joints, the synovium, is a source of stem cells for the repair of articular cartilage defects. We investigated the potential of the normal human synovium to form novel cartilage and compared its chondrogenic capacity with that of two patient groups suffering from major joint diseases: young adults with femoro-acetabular impingement syndromes of the hip (FAI), and elderly individuals with osteoarthritic degeneration of the knee (OA). Synovial membrane explants of these three patient groups were induced in vitro to undergo chondrogenesis by growth factors: bone morphogenetic protein-2 (BMP-2) alone, transforming growth factor-β1 (TGF-β1) alone, or a combination of these two. Quantitative evaluations of the newly formed cartilages were performed respecting their gene activities, as well as the histochemical, immunhistochemical, morphological and histomorphometrical characteristics. Formation of adult articular-like cartilage was induced by the BMP-2/TGF-β1 combination within all three groups, and was confirmed by adequate gene-expression levels of the anabolic chondrogenic markers; the levels of the catabolic markers remained low. Our data reveal that the chondrogenic potential of the normal human synovium remains uncompromised, both in FAI and OA. The potential of synovium-based clinical repair of joint cartilage may thus not be impaired by age-related joint pathologies.
Collapse
Affiliation(s)
- Ernst B Hunziker
- Departments of Osteoporosis and Orthopaedic Surgery, Inselspital Bern University Hospital, Freiburgstrasse 3, 3010, Bern, Switzerland.
| | - Nahoko Shintani
- Department of Osteoporosis, Inselspital Bern University Hospital, Bern, Switzerland
| | - Kurt Lippuner
- Department of Osteoporosis, Inselspital Bern University Hospital, Bern, Switzerland
| | - Esther Vögelin
- Departments of Plastic and Hand Surgery, Inselspital Bern University Hospital, Bern, Switzerland
| | - Marius J B Keel
- Trauma Center Hirslanden, Clinic Hirslanden, Zurich, Switzerland
- Medical School, University of Zurich, Zurich, Switzerland
- Department of Orthopaedic Surgery, Inselspital Bern University Hospital, Bern, Switzerland
| |
Collapse
|
12
|
Cruz MA, Gonzalez Y, Vélez Toro JA, Karimzadeh M, Rubbo A, Morris L, Medam R, Splawn T, Archer M, Fernandes RJ, Dennis JE, Kean TJ. Micronutrient optimization for tissue engineered articular cartilage production of type II collagen. Front Bioeng Biotechnol 2023; 11:1179332. [PMID: 37346792 PMCID: PMC10280293 DOI: 10.3389/fbioe.2023.1179332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Tissue Engineering of cartilage has been hampered by the inability of engineered tissue to express native levels of type II collagen in vitro. Inadequate levels of type II collagen are, in part, due to a failure to recapitulate the physiological environment in culture. In this study, we engineered primary rabbit chondrocytes to express a secreted reporter, Gaussia Luciferase, driven by the type II collagen promoter, and applied a Design of Experiments approach to assess chondrogenic differentiation in micronutrient-supplemented medium. Using a Response Surface Model, 240 combinations of micronutrients absent in standard chondrogenic differentiation medium, were screened and assessed for type II collagen promoter-driven Gaussia luciferase expression. While the target of this study was to establish a combination of all micronutrients, alpha-linolenic acid, copper, cobalt, chromium, manganese, molybdenum, vitamins A, E, D and B7 were all found to have a significant effect on type II collagen promoter activity. Five conditions containing all micronutrients predicted to produce the greatest luciferase expression were selected for further study. Validation of these conditions in 3D aggregates identified an optimal condition for type II collagen promoter activity. Engineered cartilage grown in this condition, showed a 170% increase in type II collagen expression (Day 22 Luminescence) and in Young's tensile modulus compared to engineered cartilage in basal media alone.Collagen cross-linking analysis confirmed formation of type II-type II collagen and type II-type IX collagen cross-linked heteropolymeric fibrils, characteristic of mature native cartilage. Combining a Design of Experiments approach and secreted reporter cells in 3D aggregate culture enabled a high-throughput platform that can be used to identify more optimal physiological culture parameters for chondrogenesis.
Collapse
Affiliation(s)
- Maria A. Cruz
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Yamilet Gonzalez
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Javier A. Vélez Toro
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Makan Karimzadeh
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Anthony Rubbo
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Lauren Morris
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Ramapaada Medam
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Taylor Splawn
- Baylor College of Medicine, Houston, TX, United States
| | - Marilyn Archer
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, United States
| | - Russell J. Fernandes
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, United States
| | | | - Thomas J. Kean
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
- Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
13
|
Fainor M, Mahindroo S, Betz KR, Augustin J, Smith HE, Mauck RL, Gullbrand SE. A Tunable Calcium Phosphate Coating to Drive in vivo Osseointegration of Composite Engineered Tissues. Cells Tissues Organs 2023; 212:383-398. [PMID: 36966531 PMCID: PMC10616759 DOI: 10.1159/000528965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/30/2022] [Indexed: 06/18/2023] Open
Abstract
Varying degrees of hydroxyapatite (HA) surface functionalization have been implicated as the primary driver of differential osteogenesis observed in infiltrating cells. The ability to reliably create spatially controlled areas of mineralization in composite engineered tissues is of growing interest in the field, and the use of HA-functionalized biomaterials may provide a robust solution to this challenge. In this study, we successfully fabricated polycaprolactone salt-leached scaffolds with two levels of a biomimetic calcium phosphate coating to examine their effects on MSC osteogenesis. Longer duration coating in simulated body fluid (SBF) led to increased HA crystal nucleation within scaffold interiors as well as more robust HA crystal formation on scaffold surfaces. Ultimately, the increased surface stiffness of scaffolds coated in SBF for 7 days in comparison to scaffolds coated in SBF for 1 day led to more robust osteogenesis of MSCs in vitro without the assistance of osteogenic signaling molecules. This study also demonstrated that the use of SBF-based HA coatings can promote higher levels of osteogenesis in vivo. Finally, when incorporated as the endplate region of a larger tissue-engineered intervertebral disc replacement, HA coating did not induce mineralization in or promote cell migration out of neighboring biomaterials. Overall, these results verified tunable biomimetic HA coatings as a promising biomaterial modification to promote discrete regions of mineralization within composite engineered tissues.
Collapse
Affiliation(s)
- Matthew Fainor
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, (PA,) USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
| | - Sonal Mahindroo
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
- Department of Biology, St. Bonaventure University, St. Bonaventure, (NY,) USA
| | - Kerri R. Betz
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
- Department of Biology, Widener University, Chester, (PA,) USA
| | - Janai Augustin
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
- Department of Biomedical Engineering, City College of New York, New York City, (NY,) USA
| | - Harvey E. Smith
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, (PA,) USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
| | - Robert L. Mauck
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, (PA,) USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, (PA,) USA
| | - Sarah E. Gullbrand
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, (PA,) USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
| |
Collapse
|
14
|
Donnelly H, Kurjan A, Yong LY, Xiao Y, Lemgruber L, West C, Salmeron-Sanchez M, Dalby MJ. Fibronectin matrix assembly and TGFβ1 presentation for chondrogenesis of patient derived pericytes for microtia repair. BIOMATERIALS ADVANCES 2023; 148:213370. [PMID: 36931082 DOI: 10.1016/j.bioadv.2023.213370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/10/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
Tissue engineered cartilage for external ear reconstruction of congenital deformities, such as microtia or resulting from trauma, remains a significant challenge for plastic and reconstructive surgeons. Current strategies involve harvesting autologous costal cartilage or expanding autologous chondrocytes ex vivo. However, these procedures often lead to donor site morbidity and a cell source with limited expansion capacity. Stromal stem cells such as perivascular stem cells (pericytes) offer an attractive alternative cell source, as they can be isolated from many human tissues, readily expanded in vitro and possess chondrogenic differentiation potential. Here, we successfully isolate CD146+ pericytes from the microtia remnant from patients undergoing reconstructive surgery (Microtia pericytes; MPs). Then we investigate their chondrogenic potential using the polymer poly(ethyl acrylate) (PEA) to unfold the extracellular matrix protein fibronectin (FN). FN unfolding exposes key growth factor (GF) and integrin binding sites on the molecule, allowing tethering of the chondrogenic GF transforming growth factor beta 1 (TGFβ1). This system leads to solid-phase, matrix-bound, GF presentation in a more physiological-like manner than that of typical chondrogenic induction media (CM) formulations that tend to lead to off-target effects. This simple and controlled material-based approach demonstrates similar chondrogenic potential to CM, while minimising proclivity toward hypertrophy, without the need for complex induction media formulations.
Collapse
Affiliation(s)
- Hannah Donnelly
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, United Kingdom.
| | - Alina Kurjan
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Li Yenn Yong
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Yinbo Xiao
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Leandro Lemgruber
- Glasgow Imaging Facility, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Christopher West
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
15
|
Wuttisiriboon K, Tippayawat P, Daduang J, Limpaiboon T. Ca 2+/Calmodulin-dependent Protein Kinase II Inhibitor KN-93 Enhances Chondrogenesis of Bone Marrow Mesenchymal Stem Cells and Delays Chondrogenic Hypertrophy. In Vivo 2023; 37:667-678. [PMID: 36881077 PMCID: PMC10026659 DOI: 10.21873/invivo.13127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/04/2023] [Accepted: 02/09/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND/AIM Cartilage tissue engineering has been popularly applied in the treatment of articular cartilage defect because it is more effective in generating functional engineered cartilage than traditional methods. Although the chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells (BM-MSCs) is well established, it is often accompanied by undesired hypertrophy. Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a crucial mediator in the ion channel pathway which is known to be involved in chondrogenic hypertrophy. Therefore, this study aimed to reduce the hypertrophy of BM-MSCs by inhibiting CaMKII activation. MATERIALS AND METHODS BM-MSCs were cultured in three-dimensional (3D) scaffold under chondrogenic induction with and without CaMKII inhibitor, KN-93. After cultivation, markers of chondrogenesis and hypertrophy were investigated. RESULTS KN-93 at a concentration of 2.0 μM had no effect on the viability of BM-MSCs, while the activation of CaMKII was suppressed. A long period of KN-93 treatment significantly up-regulated the expression of SRY-box transcription factor 9 and aggrecan on day 28 compared to untreated BM-MSCs. Furthermore, KN-93 treatment significantly down-regulated the expression of RUNX family transcription factor 2 and collagen type X alpha 1 chain on days 21 and 28. Immunohistochemistry showed increased expression of aggrecan and type II collagen while the expression of type X collagen was reduced. CONCLUSION A CaMKII inhibitor, KN-93 is able to enhance chondrogenesis of BM-MSCs and suppress chondrogenic hypertrophy, suggesting its potential applicability in cartilage tissue engineering.
Collapse
Affiliation(s)
| | - Patcharaporn Tippayawat
- School of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Jureerut Daduang
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Temduang Limpaiboon
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
16
|
Ong LJY, Fan X, Rujia Sun A, Mei L, Toh YC, Prasadam I. Controlling Microenvironments with Organs-on-Chips for Osteoarthritis Modelling. Cells 2023; 12:579. [PMID: 36831245 PMCID: PMC9954502 DOI: 10.3390/cells12040579] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Osteoarthritis (OA) remains a prevalent disease affecting more than 20% of the global population, resulting in morbidity and lower quality of life for patients. The study of OA pathophysiology remains predominantly in animal models due to the complexities of mimicking the physiological environment surrounding the joint tissue. Recent development in microfluidic organ-on-chip (OoC) systems have demonstrated various techniques to mimic and modulate tissue physiological environments. Adaptations of these techniques have demonstrated success in capturing a joint tissue's tissue physiology for studying the mechanism of OA. Adapting these techniques and strategies can help create human-specific in vitro models that recapitulate the cellular processes involved in OA. This review aims to comprehensively summarise various demonstrations of microfluidic platforms in mimicking joint microenvironments for future platform design iterations.
Collapse
Affiliation(s)
- Louis Jun Ye Ong
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane City, QLD 4000, Australia
| | - Xiwei Fan
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Antonia Rujia Sun
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Lin Mei
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Centre for Microbiome Research, Queensland University of Technology, Brisbane City, QLD 4000, Australia
| | - Indira Prasadam
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| |
Collapse
|
17
|
Stoddart MJ, Della Bella E, Armiento AR. Cartilage Tissue Engineering: An Introduction. Methods Mol Biol 2023; 2598:1-7. [PMID: 36355280 DOI: 10.1007/978-1-0716-2839-3_1] [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: 06/16/2023]
Abstract
Once damaged, cartilage has limited healing capability. This has led to a huge body of research that aims to repair or regenerate this important tissue. Despite the progress made, significant hurdles still need to be overcome. This chapter highlights some of the progress made, while elaborating on areas that need further research. The concept of translation and the route to clinical translation must be kept in mind if some of the promising preclinical research is to make it to routine clinical application.
Collapse
Affiliation(s)
| | | | - Angela R Armiento
- AO Research Institute Davos, Davos Platz, Switzerland
- UCB Pharma, Slough, UK
| |
Collapse
|
18
|
Salthouse D, Novakovic K, Hilkens CMU, Ferreira AM. Interplay between biomaterials and the immune system: Challenges and opportunities in regenerative medicine. Acta Biomater 2023; 155:1-18. [PMID: 36356914 DOI: 10.1016/j.actbio.2022.11.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/17/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
Abstract
The use of biomaterials for tissue engineering and regenerative medicine applications has increased dramatically over recent years. However, the clinical uptake of a wide variety of biomaterials remains limited due to adverse effects commonly exhibited by patients, which are caused by the host immune response. Despite this, current in vitro evaluation standards (ISO-10993) for assessing the host response to biomaterials have limitations in predicting the likelihood of in vivo biomaterial acceptance. Furthermore, endotoxin contamination of biomaterials is rarely considered, despite its ability to significantly affect the performance of biomaterials and engineered tissues. This review highlights the importance of the immune response to biomaterials and discusses existing challenges and opportunities in the development and standardised assessment of the immune response to biomaterials, including the importance of endotoxin levels. In addition, the properties of biomaterials that impact the host immune response and the exploitation of immunomodulatory biomaterials in regenerative medicine are explored. Finally, a standardised in vitro pathway of evaluating the immune response to biomaterials (hydrogels) and their regenerative potential is proposed, aiming to ensure safety and consistency, while reducing costs and the use of animals in the biomaterials research for tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE: This review presents a critical analysis of the role of the interactions between the immune system and biomaterials in determining the therapeutic success of biomaterial-based approaches. No such review addressing the lack of understanding of biomaterial-immune system interactions during the developmental and pre-clinical stages of biomaterials, including the impact of the endotoxin levels of biomaterials on the immune response, is published. As there is a lack of in vitro regulations to evaluate the immune response to biomaterials, a standardised in vitro pathway to evaluate the immune response to biomaterials (hydrogels) and their immunomodulatory and regenerative potential for use in tissue engineering/regenerative medicine applications is presented. The aim of the proposed pathway of biomaterial evaluation is to ensure safety and consistency in the biomaterials research community, while reducing costs and animal use (through the concept of the 3R's - reduction, refinement, and replacement of animals).
Collapse
Affiliation(s)
- Daniel Salthouse
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Katarina Novakovic
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Catharien M U Hilkens
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom.
| |
Collapse
|
19
|
Brose TZ, Ladner YD, Kubosch EJ, Stoddart MJ, Armiento AR. Co-culture of Human Articular Chondrocytes Seeded in Polyurethane Scaffolds and Human Mesenchymal Stromal Cells Encapsulated in Alginate Beads. Methods Mol Biol 2023; 2598:177-186. [PMID: 36355292 DOI: 10.1007/978-1-0716-2839-3_13] [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: 06/16/2023]
Abstract
Co-culturing is an essential method for unravelling the importance of cross talk and cellular interaction. This chapter describes the preparation of an indirect co-culture technique based on encapsulation of chondrocytes and mesenchymal stromal cells in polyurethane scaffolds and alginate beads, respectively. This way, both cell populations can communicate through paracrine effects in the absence of cell-cell contact. Due to the mechanical properties of polyurethane, this model can be employed in mechanobiology studies. The resulting engineered cultures can provide a more realistic environment, recreating the complex joints' microenvironment and physiology.
Collapse
Affiliation(s)
- Teresa Z Brose
- Medical Center-Albert-Ludwigs-University, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Yann D Ladner
- Regenerative Orthopaedics Program, AO Research Institute Davos, Davos Platz, Switzerland
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - E Johanna Kubosch
- Medical Center-Albert-Ludwigs-University, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Martin J Stoddart
- Regenerative Orthopaedics Program, AO Research Institute Davos, Davos Platz, Switzerland
| | - Angela R Armiento
- Regenerative Orthopaedics Program, AO Research Institute Davos, Davos Platz, Switzerland.
- UCB Pharma, Slough, UK.
| |
Collapse
|
20
|
Esmaeili A, Hosseini S, Kamali A, Hosseinzadeh M, Shekari F, Baghaban Eslaminejad M. Co-aggregation of MSC/chondrocyte in a dynamic 3D culture elevates the therapeutic effect of secreted extracellular vesicles on osteoarthritis in a rat model. Sci Rep 2022; 12:19827. [PMID: 36400827 PMCID: PMC9674636 DOI: 10.1038/s41598-022-22592-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
Extracellular vesicles (EVs) have therapeutic effects on osteoarthritis (OA). Some recent strategies could elevate EV's therapeutic properties including cell aggregation, co-culture, and 3D culture. It seems that a combination of these strategies could augment EV production and therapeutic potential. The current study aims to evaluate the quantity of EV yield and the therapeutic effect of EVs harvested from rabbit mesenchymal stem cells (MSCs) aggregates, chondrocyte aggregates, and their co-aggregates in a dynamic 3D culture in a rat osteoarthritis model. MSC and chondrocytes were aggregated and co-aggregated by spinner flasks, and their conditioned medium was collected. EVs were isolated by size exclusion chromatography and characterized in terms of size, morphology and surface markers. The chondrogenic potential of the MSC-ag, Cho-ag and Co-ag EVs on MSC micromass differentiation in chondrogenic media were assessed by qRT-PCR, histological and immunohistochemical analysis. 50 μg of MSC-ag-EVs, Cho-ag-EVs and Co-ag-EVs was injected intra-articularly per knee of OA models established by monoiodoacetate in rats. After 8 weeks follow up, the knee joints were harvested and analyzed by radiographic, histological and immunohistochemical features. MSC/chondrocyte co-aggregation in comparison to MSC or chondrocyte aggregation could increase EV yield during dynamic 3D culture by spinner flasks. Although MSC-ag-, Cho-ag- and Co-ag-derived EVs could induce chondrogenesis similar to transforming growth factor-beta during in vitro study, Co-ag-EV could more effectively prevent OA progression than MSC-ag- and Cho-ag-EVs. Our study demonstrated that EVs harvested from the co-aggregation of MSCs and chondrocytes could be considered as a new therapeutic potential for OA treatment.
Collapse
Affiliation(s)
- Abazar Esmaeili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Kamali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Hosseinzadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
21
|
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
|
22
|
Spatial patterning of phenotypically distinct microtissues to engineer osteochondral grafts for biological joint resurfacing. Biomaterials 2022; 289:121750. [DOI: 10.1016/j.biomaterials.2022.121750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/11/2022] [Accepted: 08/16/2022] [Indexed: 02/04/2023]
|
23
|
Wu S, Guo W, Li R, Zhang X, Qu W. Progress of Platelet Derivatives for Cartilage Tissue Engineering. Front Bioeng Biotechnol 2022; 10:907356. [PMID: 35782516 PMCID: PMC9243565 DOI: 10.3389/fbioe.2022.907356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Articular cartilage has limited self-regeneration ability for lacking of blood vessels, nerves, and lymph that makes it a great challenge to repair defects of the tissue and restore motor functions of the injured or aging population. Platelet derivatives, such as platelet-rich plasma, have been proved effective, safe, and economical in musculoskeletal diseases for their autologous origin and rich in growth factors. The combination of platelet derivatives with biomaterials provides both mechanical support and localized sustained release of bioactive molecules in cartilage tissue engineering and low-cost efficient approaches of potential treatment. In this review, we first provide an overview of platelet derivatives and their application in clinical and experimental therapies, and then we further discuss the techniques of the addition of platelet derivatives and their influences on scaffold properties. Advances in cartilage tissue engineering with platelet derivatives as signal factors and structural components are also introduced before prospects and concerns in this research field. In short, platelet derivatives have broad application prospects as an economical and effective enhancement for tissue engineering–based articular cartilage repair.
Collapse
Affiliation(s)
- Siyu Wu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Wenlai Guo
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Rui Li
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xi Zhang
- Department of Burn Surgery, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xi Zhang, ; Wenrui Qu,
| | - Wenrui Qu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Xi Zhang, ; Wenrui Qu,
| |
Collapse
|
24
|
Contentin R, Jammes M, Bourdon B, Cassé F, Bianchi A, Audigié F, Branly T, Velot É, Galéra P. Bone Marrow MSC Secretome Increases Equine Articular Chondrocyte Collagen Accumulation and Their Migratory Capacities. Int J Mol Sci 2022; 23:5795. [PMID: 35628604 PMCID: PMC9146805 DOI: 10.3390/ijms23105795] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
Equine osteoarthritis (OA) leads to cartilage degradation with impaired animal well-being, premature cessation of sport activity, and financial losses. Mesenchymal stem cell (MSC)-based therapies are promising for cartilage repair, but face limitations inherent to the cell itself. Soluble mediators and extracellular vesicles (EVs) secreted by MSCs are the alternatives to overcome those limitations while preserving MSC restorative properties. The effect of equine bone marrow MSC secretome on equine articular chondrocytes (eACs) was analyzed with indirect co-culture and/or MSC-conditioned media (CM). The expression of healthy cartilage/OA and proliferation markers was evaluated in eACs (monolayers or organoids). In vitro repair experiments with MSC-CM were made to evaluate the proliferation and migration of eACs. The presence of nanosized EVs in MSC-CM was appraised with nanoparticle tracking assay and transmission electron microscopy. Our results demonstrated that the MSC secretome influences eAC phenotype by increasing cartilage functionality markers and cell migration in a greater way than MSCs, which could delay OA final outcomes. This study makes acellular therapy an appealing strategy to improve equine OA treatments. However, the MSC secretome contains a wide variety of soluble mediators and small EVs, such as exosomes, and further investigation must be performed to understand the mechanisms occurring behind these promising effects.
Collapse
Affiliation(s)
- Romain Contentin
- Normandie University, Unicaen, Biotargen, F-14000 Caen, France; (R.C.); (M.J.); (B.B.); (F.C.); (T.B.)
| | - Manon Jammes
- Normandie University, Unicaen, Biotargen, F-14000 Caen, France; (R.C.); (M.J.); (B.B.); (F.C.); (T.B.)
| | - Bastien Bourdon
- Normandie University, Unicaen, Biotargen, F-14000 Caen, France; (R.C.); (M.J.); (B.B.); (F.C.); (T.B.)
| | - Frédéric Cassé
- Normandie University, Unicaen, Biotargen, F-14000 Caen, France; (R.C.); (M.J.); (B.B.); (F.C.); (T.B.)
| | - Arnaud Bianchi
- Molecular Engineering and Articular Physiopathology (IMoPA), French National Center for Scientific Research (CNRS), Université de Lorraine, F-54000 Nancy, France; (A.B.); (É.V.)
| | - Fabrice Audigié
- Center of Imaging and Research on Locomotor Affections on Equines (CIRALE), Unit Under Contract 957 Equine Biomechanics and Locomotor Disorders (USC 957 BPLC), French National Research Institute for Agriculture Food and Environment (INRAE), École Nationale Vétérinaire d’Alfort, F-94700 Maisons-Alfort, France;
| | - Thomas Branly
- Normandie University, Unicaen, Biotargen, F-14000 Caen, France; (R.C.); (M.J.); (B.B.); (F.C.); (T.B.)
| | - Émilie Velot
- Molecular Engineering and Articular Physiopathology (IMoPA), French National Center for Scientific Research (CNRS), Université de Lorraine, F-54000 Nancy, France; (A.B.); (É.V.)
| | - Philippe Galéra
- Normandie University, Unicaen, Biotargen, F-14000 Caen, France; (R.C.); (M.J.); (B.B.); (F.C.); (T.B.)
| |
Collapse
|
25
|
Biodegradable Poly(D-L-lactide-co-glycolide) (PLGA)-Infiltrated Bioactive Glass (CAR12N) Scaffolds Maintain Mesenchymal Stem Cell Chondrogenesis for Cartilage Tissue Engineering. Cells 2022; 11:cells11091577. [PMID: 35563883 PMCID: PMC9100331 DOI: 10.3390/cells11091577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 12/11/2022] Open
Abstract
Regeneration of articular cartilage remains challenging. The aim of this study was to increase the stability of pure bioactive glass (BG) scaffolds by means of solvent phase polymer infiltration and to maintain cell adherence on the glass struts. Therefore, BG scaffolds either pure or enhanced with three different amounts of poly(D-L-lactide-co-glycolide) (PLGA) were characterized in detail. Scaffolds were seeded with primary porcine articular chondrocytes (pACs) and human mesenchymal stem cells (hMSCs) in a dynamic long-term culture (35 days). Light microscopy evaluations showed that PLGA was detectable in every region of the scaffold. Porosity was greater than 70%. The biomechanical stability was increased by polymer infiltration. PLGA infiltration did not result in a decrease in viability of both cell types, but increased DNA and sulfated glycosaminoglycan (sGAG) contents of hMSCs-colonized scaffolds. Successful chondrogenesis of hMSC-colonized scaffolds was demonstrated by immunocytochemical staining of collagen type II, cartilage proteoglycans and the transcription factor SOX9. PLGA-infiltrated scaffolds showed a higher relative expression of cartilage related genes not only of pAC-, but also of hMSC-colonized scaffolds in comparison to the pure BG. Based on the novel data, our recommendation is BG scaffolds with single infiltrated PLGA for cartilage tissue engineering.
Collapse
|
26
|
Lesage C, Lafont M, Guihard P, Weiss P, Guicheux J, Delplace V. Material-Assisted Strategies for Osteochondral Defect Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200050. [PMID: 35322596 PMCID: PMC9165504 DOI: 10.1002/advs.202200050] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/25/2022] [Indexed: 05/08/2023]
Abstract
The osteochondral (OC) unit plays a pivotal role in joint lubrication and in the transmission of constraints to bones during movement. The OC unit does not spontaneously heal; therefore, OC defects are considered to be one of the major risk factors for developing long-term degenerative joint diseases such as osteoarthritis. Yet, there is currently no curative treatment for OC defects, and OC regeneration remains an unmet medical challenge. In this context, a plethora of tissue engineering strategies have been envisioned over the last two decades, such as combining cells, biological molecules, and/or biomaterials, yet with little evidence of successful clinical transfer to date. This striking observation must be put into perspective with the difficulty in comparing studies to identify overall key elements for success. This systematic review aims to provide a deeper insight into the field of material-assisted strategies for OC regeneration, with particular considerations for the therapeutic potential of the different approaches (with or without cells or biological molecules), and current OC regeneration evaluation methods. After a brief description of the biological complexity of the OC unit, the recent literature is thoroughly analyzed, and the major pitfalls, emerging key elements, and new paths to success are identified and discussed.
Collapse
Affiliation(s)
- Constance Lesage
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
- HTL Biotechnology7 Rue Alfred KastlerJavené35133France
| | - Marianne Lafont
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| | - Pierre Guihard
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| | - Pierre Weiss
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| | - Jérôme Guicheux
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| | - Vianney Delplace
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| |
Collapse
|
27
|
Shen K, Duan A, Cheng J, Yuan T, Zhou J, Song H, Chen Z, Wan B, Liu J, Zhang X, Zhang Y, Xie R, Liu F, Fan W, Zuo Q. Exosomes derived from hypoxia preconditioned mesenchymal stem cells laden in a silk hydrogel promote cartilage regeneration via the miR-205-5p/PTEN/AKT pathway. Acta Biomater 2022; 143:173-188. [PMID: 35202856 DOI: 10.1016/j.actbio.2022.02.026] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022]
Abstract
Tissue engineering has promising prospects for cartilage regeneration. However, there remains an urgent need to harvest high quality seed cells. Bone marrow mesenchymal cells (BMSCs), and in particular their exosomes, might promote the function of articular chondrocytes (ACs) via paracrine mechanisms. Furthermore, preconditioned BMSCs could provide an enhanced therapeutic effect. BMSCs naturally exist in a relatively hypoxic environment (1%-5% O2); however, they are usually cultured under higher oxygen concentrations (21% O2). Herein, we hypothesized that hypoxia preconditioned exosomes (H-Exos) could improve the quality of ACs and be more conducive to cartilage repair. In our study, we compared the effects of exosomes derived from BMSCs preconditioned with hypoxia and normoxia (N-Exos) on ACs, demonstrating that H-Exos significantly promoted the proliferation, migration, anabolism and anti-inflammation effects of ACs. Furthermore, we confirmed that hypoxia preconditioning upregulated the expression of miR-205-5p in H-Exos, suggesting that ACs were promoted via the miR-205-5p/PTEN/AKT pathway. Finally, an injectable silk fibroin (SF) hydrogel containing ACs and H-Exos (SF/ACs/H-Exos) was utilized to repair cartilage defects and effectively promote cartilage regeneration in vivo. The application of SF/ACs/H-Exos hydrogel in cartilage regeneration therefore has promising prospects. STATEMENT OF SIGNIFICANCE: Cartilage tissue engineering (CTE) has presented a promising prospect. However, the quality of seed cells is an important factor affecting the repair efficiency. Our study demonstrates for the first time that the exosomes derived from hypoxia preconditioned BMSCs (H-Exos) effectively promote the proliferation, migration and anabolism of chondrocytes and inhibit inflammation through miR-205-5p/PTEN/AKT pathway. Furthermore, we fabricated an injectable silk fibrion (SF) hydrogel to preserve and sustained release H-Exos. A complex composed of SF hydrogel, H-Exos and chondrocytes can effectively promote the regeneration of cartilage defects. Therefore, this study demonstrates that hypoxia pretreatment could optimize the therapeutic effects of BMSCs-derived exosomes, and the combination of exosomes and SF hydrogel could be a promising therapeutic method for cartilage regeneration.
Collapse
|
28
|
Esmaeili A, Alini M, Baghaban Eslaminejad M, Hosseini S. Engineering strategies for customizing extracellular vesicle uptake in a therapeutic context. Stem Cell Res Ther 2022; 13:129. [PMID: 35346367 PMCID: PMC8960087 DOI: 10.1186/s13287-022-02806-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/07/2022] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are advanced therapeutic strategies that can be used to efficiently treat diseases. Promising features of EVs include their innate therapeutic properties and ability to be engineered as targeted drug delivery systems. However, regulation of EV uptake is one challenge of EV therapy that must be overcome to achieve an efficient therapeutic outcome. Numerous efforts to improve the factors that affect EV uptake include the selection of a cell source, cell cultivation procedure, extraction and purification methods, storage, and administration routes. Limitations of rapid clearance, targeted delivery, and off-targeting of EVs are current challenges that must be circumvented. EV engineering can potentially overcome these limitations and provide an ideal therapeutic use for EVs. In this paper, we intend to discuss traditional strategies and their limitations, and then review recent advances in EV engineering that can be used to customize and control EV uptake for future clinical applications.
Collapse
Affiliation(s)
- Abazar Esmaeili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
29
|
Posniak S, Chung JHY, Liu X, Mukherjee P, Gambhir S, Khansari A, Wallace GG. Bioprinting of Chondrocyte Stem Cell Co-Cultures for Auricular Cartilage Regeneration. ACS OMEGA 2022; 7:5908-5920. [PMID: 35224351 PMCID: PMC8867593 DOI: 10.1021/acsomega.1c06102] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/21/2022] [Indexed: 05/06/2023]
Abstract
Advances in 3D bioprinting allows not only controlled deposition of cells or cell-laden hydrogels but also flexibility in creating constructs that match the anatomical features of the patient. This is especially the case for reconstructing the pinna (ear), which is a large feature of the face and made from elastic cartilage that primarily relies on diffusion for nutrient transfer. The selection of cell lines for reconstructing this cartilage becomes a crucial step in clinical translation. Chondrocytes and mesenchymal stem cells are both studied extensively in the area of cartilage regeneration as they are capable of producing cartilage in vitro. However, such monoculture systems involve unfavorable processes and produce cartilage with suboptimal characteristics. Co-cultures of these cell types are known to alleviate these limitations to produce synergically active chondrocytes and cartilage. The current study utilized a 3D bioprinted scaffold made from combined gelatine methacryloyl and methacrylated hyaluronic acid (GelMA/HAMA) to interrogate monocultures and co-cultures of human septal chondrocytes (primary chondrocytes, PCs) and human bone marrow-derived mesenchymal stem cells (BM-hMSCs). This study is also the first to examine co-cultures of healthy human chondrocytes with human BM-hMSCs encapsulated in GelMA/HAMA bioprinted scaffolds. Findings revealed that the combination of MSCs and PCs not only yielded cell proliferation that mimicked MSCs but also produced chondrogenic expressions that mimicked PCs. These findings suggested that co-cultures of BM-hMSCs and healthy septal PCs can be employed to replace monocultures in chondrogenic studies for cartilage regeneration in this model. The opportunity for MSCs used to replace PCs alleviates the requirement of large cartilage biopsies that would otherwise be needed for sufficient cell numbers and therefore can be employed for clinical applications.
Collapse
Affiliation(s)
- Steven Posniak
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Johnson H. Y. Chung
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Xiao Liu
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Payal Mukherjee
- ENT
Research Lead, RPA Institute of Academic Surgery, Sydney Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia
| | - Sanjeev Gambhir
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Afsaneh Khansari
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Gordon G. Wallace
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| |
Collapse
|
30
|
Recent Developments in Hyaluronic Acid-Based Hydrogels for Cartilage Tissue Engineering Applications. Polymers (Basel) 2022; 14:polym14040839. [PMID: 35215752 PMCID: PMC8963043 DOI: 10.3390/polym14040839] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 01/27/2023] Open
Abstract
Articular cartilage lesions resulting from injurious impact, recurring loading, joint malalignment, etc., are very common and encompass the risk of evolving to serious cartilage diseases such as osteoarthritis. To date, cartilage injuries are typically treated via operative procedures such as autologous chondrocyte implantation (ACI), matrix-associated autologous chondrocyte implantation (MACI) and microfracture, which are characterized by low patient compliance. Accordingly, cartilage tissue engineering (CTE) has received a lot of interest. Cell-laden hydrogels are favorable candidates for cartilage repair since they resemble the native tissue environment and promote the formation of extracellular matrix. Various types of hydrogels have been developed so far for CTE applications based on both natural and synthetic biomaterials. Among these materials, hyaluronic acid (HA), a principal component of the cartilage tissue which can be easily modified and biofunctionalized, has been favored for the development of hydrogels since it interacts with cell surface receptors, supports the growth of chondrocytes and promotes the differentiation of mesenchymal stem cells to chondrocytes. The present work reviews the various types of HA-based hydrogels (e.g., in situ forming hydrogels, cryogels, microgels and three-dimensional (3D)-bioprinted hydrogel constructs) that have been used for cartilage repair, specially focusing on the results of their preclinical and clinical assessment.
Collapse
|
31
|
Marchan J, Wittig O, Diaz-Solano D, Gomez M, Cardier JE. Enhanced chondrogenesis from chondrocytes co-cultured on mesenchymal stromal cells: Implication for cartilage repair. Injury 2022; 53:399-407. [PMID: 34670674 DOI: 10.1016/j.injury.2021.09.061] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/17/2021] [Accepted: 09/28/2021] [Indexed: 02/02/2023]
Abstract
Cellular therapy based on chondrocytes implantation is the most widely used procedure for inducing cartilage regeneration. However, the dedifferentiation process that these cells suffer and their limited capacity of proliferation, when they are cultured in vitro, restrict their use in cellular therapy protocols. To investigate the capacity of mesenchymal stromal cells (MSCs) to promote chondrogenesis from chondrocytes or chondrons in 2D and 3D coculture systems. Murine chondrocytes and chondrons were cocultured with MSCs at different cell ratios (100/0, 50/50, 70/30, 0/100) in two-dimensional (2D) and three-dimensional (3D) culture systems. High proliferation of cells with chondrocyte morphology, enhanced GAG production and expression of cartilage genes (aggrecan, type II collagen, and SOX-9) were observed in chondrocytes/MSCs cocultures. In contrast, fibroblastoid cells, down-regulation of cartilage gene expression and reduction of GAG production were observed in chondrons/MSCs cocultures. Chondrocytes within cartilage lacunae and surrounded by extracellular matrix were observed in chondrocytes/MSC pellets. MSCs promote the proliferation of functional chondrocytes in 2D and 3D culture systems. Transplantation of chondrogenic construct based on MSCs and chondrocytes may constitute a potential treatment for inducing cartilage repair.
Collapse
Affiliation(s)
- Jose Marchan
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela.
| | - Olga Wittig
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela.
| | - Dylana Diaz-Solano
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Marcos Gomez
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Jose E Cardier
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| |
Collapse
|
32
|
Chen K, Chen H, Gao H, Zhou W, Zheng S, Chen Y, Zhang S, Yao Y. Effect of passage number of genetically modified TGF-β3 expressing primary chondrocytes on the chondrogenesis of ATDC5 cells in a 3D coculture system. Biomed Mater 2022; 17. [DOI: 10.1088/1748-605x/ac489e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/06/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Due to the lack of blood vessels, nerves and lymphatics, articular cartilage is difficult to repair once damaged. Tissue engineering is considered to be a potential strategy for cartilage regeneration. Successful tissue engineering strategies depend on the effective combination of biomaterials, seed cells and biological factors. In our previous study, a genetically modified coculture system with chondrocytes and ATDC5 cells in an alginate hydrogel has exhibited a superior ability to enhance chondrogenesis. In this study, we further evaluated the influence of chondrocytes at various passages on chondrogenesis in the coculture system. The results demonstrated that transfection efficiency was hardly influenced by the passage of chondrocytes. The coculture system with passage 5 (P5) chondrocytes had a better effect on chondrogenesis of ATDC 5 cells, while chondrocytes in this coculture system presented higher levels of dedifferentiation than other groups with P1 or P3 chondrocytes. Therefore, P5 chondrocytes were shown to be more suitable for the coculture system, as they accumulated in sufficient cell numbers with more passages and had a higher level of dedifferentiation, which was prone to form a favorable niche for chondrogenesis of ATDC5 cells. This study may provide fresh insights for future cartilage tissue engineering strategies with a combination of a coculture system and advanced biomaterials.
Collapse
|
33
|
Wang B, Liu W, Li JJ, Chai S, Xing D, Yu H, Zhang Y, Yan W, Xu Z, Zhao B, Du Y, Jiang Q. A low dose cell therapy system for treating osteoarthritis: In vivo study and in vitro mechanistic investigations. Bioact Mater 2022; 7:478-490. [PMID: 34466747 PMCID: PMC8379370 DOI: 10.1016/j.bioactmat.2021.05.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be effective in alleviating the progression of osteoarthritis (OA). However, low MSC retention and survival at the injection site frequently require high doses of cells and/or repeated injections, which are not economically viable and create additional risks of complications. In this study, we produced MSC-laden microcarriers in spinner flask culture as cell delivery vehicles. These microcarriers containing a low initial dose of MSCs administered through a single injection in a rat anterior cruciate ligament (ACL) transection model of OA achieved similar reparative effects as repeated high doses of MSCs, as evaluated through imaging and histological analyses. Mechanistic investigations were conducted using a co-culture model involving human primary chondrocytes grown in monolayer, together with MSCs grown either within 3D constructs or as a monolayer. Co-culture supernatants subjected to secretome analysis showed significant decrease of inflammatory factors in the 3D group. RNA-seq of co-cultured MSCs and chondrocytes using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed processes relating to early chondrogenesis and increased extracellular matrix interactions in MSCs of the 3D group, as well as phenotypic maintenance in the co-cultured chondrocytes. The cell delivery platform we investigated may be effective in reducing the cell dose and injection frequency required for therapeutic applications.
Collapse
Affiliation(s)
- Bin Wang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Department of Orthopaedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 10081, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Senlin Chai
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Hongsheng Yu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenjin Yan
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Zhihong Xu
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Bin Zhao
- Department of Orthopaedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| |
Collapse
|
34
|
Tang Z, Lu Y, Zhang S, Wang J, Wang Q, Xiao Y, Zhang X. Chondrocyte secretome enriched microparticles encapsulated with the chondrocyte membrane to facilitate the chondrogenesis of BMSCs and reduce hypertrophy. J Mater Chem B 2021; 9:9989-10002. [PMID: 34874033 DOI: 10.1039/d1tb02319e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Co-culture of chondrocytes and mesenchymal stem cells (MSCs) represents an effective way to stimulate the chondrogenesis of MSCs and reduce hypertrophy, but the limited donor site supply and the requirement of two-stage operations are among the major barriers of using autologous chondrocytes in clinical settings. With recent evidence indicating that the chondrogenic effects of the above co-culture mainly lied on the paracrine secretion, and that cell membranes also played crucial roles during the chondrocyte-MSC interaction, we fabricated a multifunctional design of "artificial chondrocytes", which consist of chondrocyte secretome enriched PLGA microparticles with the encapsulation of chondrocytes' membrane fragments. The artificial chondrocytes had shown a similar diameter and surface electrical charge to natural chondrocytes, with the preserved key chondrocyte membrane surface proteins and sustainedly released chondrogenic cytokines from the chondrocyte secretome to extend their effects in vivo. Consequently, the co-culture studies of artificial chondrocytes and bone marrow MSCs had shown the beneficial effects from both chondrocyte secretome and membrane fragments, which also synergistically facilitated the cell proliferation, chondrogenic gene expression, cartilaginous matrix production, and reduced phenotypic hypertrophy in vitro and in vivo. Together, this study has successfully developed the proof-of-concept design of "artificial chondrocytes", which could potentially conquer many major barriers of using natural chondrocytes and provided a novel synthetic-cell approach to current therapeutical strategies towards the functional regeneration of articular cartilage.
Collapse
Affiliation(s)
- Zizhao Tang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Yan Lu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Shixin Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Jing Wang
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu, Sichuan, 610059, China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| |
Collapse
|
35
|
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: 0.8] [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
|
36
|
Lindberg GCJ, Cui X, Durham M, Veenendaal L, Schon BS, Hooper GJ, Lim KS, Woodfield TBF. Probing Multicellular Tissue Fusion of Cocultured Spheroids-A 3D-Bioassembly Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2103320. [PMID: 34632729 PMCID: PMC8596109 DOI: 10.1002/advs.202103320] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Indexed: 05/02/2023]
Abstract
While decades of research have enriched the knowledge of how to grow cells into mature tissues, little is yet known about the next phase: fusing of these engineered tissues into larger functional structures. The specific effect of multicellular interfaces on tissue fusion remains largely unexplored. Here, a facile 3D-bioassembly platform is introduced to primarily study fusion of cartilage-cartilage interfaces using spheroids formed from human mesenchymal stromal cells (hMSCs) and articular chondrocytes (hACs). 3D-bioassembly of two adjacent hMSCs spheroids displays coordinated migration and noteworthy matrix deposition while the interface between two hAC tissues lacks both cells and type-II collagen. Cocultures contribute to increased phenotypic stability in the fusion region while close initial contact between hMSCs and hACs (mixed) yields superior hyaline differentiation over more distant, indirect cocultures. This reduced ability of potent hMSCs to fuse with mature hAC tissue further underlines the major clinical challenge that is integration. Together, this data offer the first proof of an in vitro 3D-model to reliably study lateral fusion mechanisms between multicellular spheroids and mature cartilage tissues. Ultimately, this high-throughput 3D-bioassembly model provides a bridge between understanding cellular differentiation and tissue fusion and offers the potential to probe fundamental biological mechanisms that underpin organogenesis.
Collapse
Affiliation(s)
- Gabriella C. J. Lindberg
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Xiaolin Cui
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Mitchell Durham
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Laura Veenendaal
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Benjamin S. Schon
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Gary J. Hooper
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Khoon S. Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Tim B. F. Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| |
Collapse
|
37
|
Urlić I, Ivković A. Cell Sources for Cartilage Repair-Biological and Clinical Perspective. Cells 2021; 10:cells10092496. [PMID: 34572145 PMCID: PMC8468484 DOI: 10.3390/cells10092496] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 01/04/2023] Open
Abstract
Cell-based therapy represents a promising treatment strategy for cartilage defects. Alone or in combination with scaffolds/biological signals, these strategies open many new avenues for cartilage tissue engineering. However, the choice of the optimal cell source is not that straightforward. Currently, various types of differentiated cells (articular and nasal chondrocytes) and stem cells (mesenchymal stem cells, induced pluripotent stem cells) are being researched to objectively assess their merits and disadvantages with respect to the ability to repair damaged articular cartilage. In this paper, we focus on the different cell types used in cartilage treatment, first from a biological scientist’s perspective and then from a clinician’s standpoint. We compare and analyze the advantages and disadvantages of these cell types and offer a potential outlook for future research and clinical application.
Collapse
Affiliation(s)
- Inga Urlić
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence: (I.U.); (A.I.)
| | - Alan Ivković
- Department of Orthopaedic Surgery, University Hospital Sveti Duh, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Clinical Medicine, University of Applied Health Sciences, 10000 Zagreb, Croatia
- Correspondence: (I.U.); (A.I.)
| |
Collapse
|
38
|
Aldrich ED, Cui X, Murphy CA, Lim KS, Hooper GJ, McIlwraith CW, Woodfield TBF. Allogeneic mesenchymal stromal cells for cartilage regeneration: A review of in vitro evaluation, clinical experience, and translational opportunities. Stem Cells Transl Med 2021; 10:1500-1515. [PMID: 34387402 PMCID: PMC8550704 DOI: 10.1002/sctm.20-0552] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/19/2021] [Accepted: 06/06/2021] [Indexed: 12/14/2022] Open
Abstract
The paracrine signaling, immunogenic properties and possible applications of mesenchymal stromal cells (MSCs) for cartilage tissue engineering and regenerative medicine therapies have been investigated through numerous in vitro, animal model and clinical studies. The emerging knowledge largely supports the concept of MSCs as signaling and modulatory cells, exerting their influence through trophic and immune mediation rather than as a cell replacement therapy. The virtues of allogeneic cells as a ready‐to‐use product with well‐defined characteristics of cell surface marker expression, proliferative ability, and differentiation capacity are well established. With clinical applications in mind, a greater focus on allogeneic cell sources is evident, and this review summarizes the latest published and upcoming clinical trials focused on cartilage regeneration adopting allogeneic and autologous cell sources. Moreover, we review the current understanding of immune modulatory mechanisms and the role of trophic factors in articular chondrocyte‐MSC interactions that offer feasible targets for evaluating MSC activity in vivo within the intra‐articular environment. Furthermore, bringing labeling and tracking techniques to the clinical setting, while inherently challenging, will be extremely informative as clinicians and researchers seek to bolster the case for the safety and efficacy of allogeneic MSCs. We therefore review multiple promising approaches for cell tracking and labeling, including both chimerism studies and imaging‐based techniques, that have been widely explored in vitro and in animal models. Understanding the distribution and persistence of transplanted MSCs is necessary to fully realize their potential in cartilage regeneration techniques and tissue engineering applications.
Collapse
Affiliation(s)
- Ellison D Aldrich
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand.,School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Xiaolin Cui
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| | - Caroline A Murphy
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| | - Khoon S Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| | - Gary J Hooper
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| | - C Wayne McIlwraith
- Orthopedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, Colorado State University, Fort Collins, Colorado, USA
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
39
|
Brose TZ, Kubosch EJ, Schmal H, Stoddart MJ, Armiento AR. Crosstalk Between Mesenchymal Stromal Cells and Chondrocytes: The Hidden Therapeutic Potential for Cartilage Regeneration. Stem Cell Rev Rep 2021; 17:1647-1665. [PMID: 33954877 DOI: 10.1007/s12015-021-10170-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Cartilage injuries following trauma create a puzzling clinical scenario. The finite reparative potential of articular cartilage is well known, and injuries are associated with an increased risk of osteoarthritis. Cell-based therapies have spotlighted chondrocytes and mesenchymal stromal cells (MSCs) as the functional unit of articular cartilage and the progenitor cells, respectively. The available clinical treatments cannot reproduce the biomechanical properties of articular cartilage and call for continuous investigations into alternative approaches. Co-cultures of chondrocytes and MSCs are an attractive in vitro system to step closer to the in vivo multicellular environment's complexity. Research on the mechanisms of interaction between both cell types will reveal essential cues to understand cartilage regeneration. This review describes the latest discoveries on these interactions, along with advantages and main challenges in vitro and in vivo. The successful clinical translation of in vitro studies requires establishing rigorous standards and clinically relevant research models and an organ-targeting therapeutic strategy.
Collapse
Affiliation(s)
- Teresa Z Brose
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.,Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Eva J Kubosch
- Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Hagen Schmal
- Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.,Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Angela R Armiento
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.
| |
Collapse
|
40
|
Improving In Vitro Cartilage Generation by Co-Culturing Adipose-Derived Stem Cells and Chondrocytes on an Allograft Adipose Matrix Framework. Plast Reconstr Surg 2021; 147:87-99. [PMID: 33002984 DOI: 10.1097/prs.0000000000007511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Microtia is an inherited condition that results in varying degrees of external ear deformities; the most extreme form is anotia. Effective surgical reconstruction techniques have been developed. However, these usually require multistage procedures and have other inherent disadvantages. Tissue engineering technologies offer new approaches in the field of external ear reconstruction. In this setting, chondrocytes are cultured in the laboratory with the aim of creating bioengineered cartilage matrices. However, cartilage engineering has many challenges, including difficulty in culturing sufficient chondrocytes. To overcome these hurdles, the authors propose a novel model of cartilage engineering that involves co-culturing chondrocytes and adipose-derived stem cells on an allograft adipose-derived extracellular matrix scaffold. METHODS Auricular chondrocytes from porcine ear were characterized. Adipose-derived stem cells were isolated and expanded from human lipoaspirate. Then, the auricular chondrocytes were cultured on the allograft adipose matrix either alone or with the adipose-derived stem cells at different ratios and examined histologically. RESULTS Cartilage induction was most prominent when the cells were co-cultured on the allograft adipose matrix at a ratio of 1:9 (auricular chondrocyte-to-adipose-derived stem cell ratio). Furthermore, because of the xenogeneic nature of the experiment, the authors were able to determine that the adipose-derived stem cells contributed to chondrogenesis by means of a paracrine stimulation of the chondrocytes. CONCLUSIONS In this situation, adipose-derived stem cells provide sufficient support to induce the formation of cartilage when the number of auricular chondrocytes available is limited. This novel model of cartilage engineering provides a setting for using the patient's own chondrocytes and adipose tissue to create a customized ear framework that could be further used for surgical reconstruction.
Collapse
|
41
|
Vinod E, Amirtham SM, Kachroo U. An assessment of bone marrow mesenchymal stem cell and human articular cartilage derived chondroprogenitor cocultures vs. monocultures. Knee 2021; 29:418-425. [PMID: 33721626 DOI: 10.1016/j.knee.2021.02.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/11/2020] [Accepted: 02/16/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Cell based therapy in cartilage repair predominantly involves the use of chondrocytes and mesenchymal stromal cells (MSC). Co-culture systems, due to their probable synergistic effect on enhancement of functional chondrogenesis and reduction in terminal differentiation have also been attempted. Chondroprogenitors, derived from articular cartilage and regarded as MSCs, have recently garnered interest for consideration in cartilage regeneration to overcome limitations associated with use of conventional cell types. The aim of this study was to assess whetherco-culturing bone marrow (BM)-MSCs and chondroprogenitors at different ratios would yield superior results in terms of surface marker expression, gene expression and chondrogenic potential. METHODS Human BM-MSCs and chondroprogenitors obtained from three osteoarthritic knee joints and subjected to monolayer expansion and pellet cultures (10,000 cells/cm2) as five test groups containing either monocultures or co-cultures (MSC: chondroprogenitors) at three different ratios (75:25, 50:50 and 25:75) were utilized. RESULTS Data analysis revealed that all groups exhibited a high expression of CD166, CD29 and CD49e. With regard to gene expression, high expression of SOX9, Aggrecan and Collagen type I; a moderate expression of Collagen type X and RUNX2; with a low expression of Collagen type II was seen. Analysis of pellet culture revealed that chondroprogenitor monoculture and chondroprogenitor dominant coculture, exhibited a subjectively larger pellet size with higher deposition of Collagen type II and glycosaminoglycan. CONCLUSION In conclusion, this study is suggestive of chondroprogenitor monoculture superiority over MSCs, either in isolation or in a coculture system and proposes further analysis of chondroprogenitors for cartilage repair.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore 632002, India; Centre for Stem Cell Research, Christian Medical College, Vellore 632002, India.
| | | | - Upasana Kachroo
- Department of Physiology, Christian Medical College, Vellore 632002, India.
| |
Collapse
|
42
|
Padalhin A, Ventura R, Kim B, Sultana T, Park CM, Lee BT. Boosting osteogenic potential and bone regeneration by co-cultured cell derived extracellular matrix incorporated porous electrospun scaffold. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:779-798. [PMID: 33375905 DOI: 10.1080/09205063.2020.1869879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Implants for bone regeneration to remedy segmental bone defects, osteomyelitis, necrotic bone tissue and non-union fractures have worldwide appeal. Although biomaterials offer most of the advantages by improving tissue growth but developments are more commonly achieved via biologically derived molecules. To aid site specific bone tissue regeneration by synthetic scaffold, cell derived extracellular matrix (ECM) can be a crucial component. In this study, co-cultured bone marrow mesenchymal stem cell and osteoblastic cells derived ECM incorporated electrospun polycaprolactone (PCL) membranes were assessed for bone tissue engineering application. The preliminary experimental details indicated that, co-culture of cells supported enhanced in vitro ECM synthesis followed by successful deposition of osteoblastic ECM into electrospun membranes. The acellular samples revealed retention of ECM related biomacromolecules (collagen, glycosaminoglycan) and partial recovery of pores after decellularization. In vitro biocompatibility tests ensured improvement of proliferation and osteoblastic differentiation of MC3T3-E1 cells in decellularized ECM containing membrane (PCL-ECM) compared to bare membrane (PCL-B) which was further confirmed by osteogenic marker proteins expression analysis. The decellularized PCL-ECM membrane allowed great improvement of bone regeneration over the bare membrane (PCL-B) in 8 mm size critical sized rat skull defects at 2 months of post implantation. In short, the outcome of this study could be impactful in development and application of cell derived ECM based synthetic electrospun templates for bone tissue engineering application.[Formula: see text].
Collapse
Affiliation(s)
- Andrew Padalhin
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Reiza Ventura
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Boram Kim
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Tamanna Sultana
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| | - Chan Mi Park
- Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| |
Collapse
|
43
|
Ahmad MR, Badar W, Ullah Khan MA, Mahmood A, Latif N, Iqbal T, Khan Assir MZ, Sleem MA. Combination of preconditioned adipose-derived mesenchymal stem cells and platelet-rich plasma improves the repair of osteoarthritis in rat. Regen Med 2020; 15:2285-2295. [PMID: 33326341 DOI: 10.2217/rme-2020-0040] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aim: To observe the combined effect of platelet-rich plasma (PRP) and preconditioned adipose-derived mesenchymal stem cells (ADMSCs) on the injured articular cartilage of the rat. Materials & methods: Animals in the study received an intra-articular injection of PRP and preconditioned ADMSCs, both in combination and separately. The response to therapeutic intervention was evaluated by inflammatory markers, proteoglycans content, chondrogenesis and gene expression analyses. Results: The combined therapy resulted in a reduction of IL-6 and TNF-α, increased proteoglycan content of the articular cartilage, upregulation of Acan, Col2a1 and PCNA genes. Downregulation of Col1a1, Col10a1 and Casp3 genes was observed as compared with the untreated osteoarthritis rat model. Conclusion: PRP potentiates the effects of ADMSCs on the repair of damaged articular cartilage.
Collapse
Affiliation(s)
- Muhammad Rauf Ahmad
- Department of Molecular Biology, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan.,Center of Excellence in Molecular Biology, University of The Punjab, Lahore, Pakistan
| | - Wafa Badar
- Faculty of Life Sciences, University of Central Punjab, Lahore, Pakistan
| | | | - Azra Mahmood
- Center of Excellence in Molecular Biology, University of The Punjab, Lahore, Pakistan
| | - Noreen Latif
- Center of Excellence in Molecular Biology, University of The Punjab, Lahore, Pakistan
| | - Tariq Iqbal
- Department of Molecular Biology, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
| | - Muhammad Zaman Khan Assir
- Department of Molecular Biology, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
| | - Mushtaq A Sleem
- Faculty of Life Sciences, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
44
|
Kim YS, Chien AJ, Guo JL, Smith BT, Watson E, Pearce HA, Koons GL, Navara AM, Lam J, Scott DW, Grande-Allen KJ, Mikos AG. Chondrogenesis of cocultures of mesenchymal stem cells and articular chondrocytes in poly(l-lysine)-loaded hydrogels. J Control Release 2020; 328:710-721. [PMID: 33010336 PMCID: PMC7749039 DOI: 10.1016/j.jconrel.2020.09.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/22/2020] [Accepted: 09/27/2020] [Indexed: 12/14/2022]
Abstract
This work investigated the effect of poly(l-lysine) (PLL) molecular weight and concentration on chondrogenesis of cocultures of mesenchymal stem cells (MSCs) and articular chondrocytes (ACs) in PLL-loaded hydrogels. An injectable dual-network hydrogel composed of a poly(N-isopropylacrylamide)-based synthetic thermogelling macromer and a chondroitin sulfate-based biological network was leveraged as a model to deliver PLL and encapsulate the two cell populations. Incorporation of PLL into the hydrogel did not affect the hydrogel's swelling properties and degradation characteristics, nor the viability of encapsulated cells. Coculture groups demonstrated higher type II collagen expression compared to the MSC monoculture group. Expression of hypertrophic phenotype was also limited in the coculture groups. Histological analysis indicated that the ratio of MSCs to ACs was an accurate predictor of the degree of long-term chondrogenesis, while the presence of PLL was shown to have a more substantial short-term effect. Altogether, this study demonstrates that coculturing MSCs with ACs can greatly enhance the chondrogenicity of the overall cell population and offers a platform to further elucidate the short- and long-term effect of polycationic factors on the chondrogenesis of MSC and AC cocultures.
Collapse
Affiliation(s)
- Yu Seon Kim
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - Athena J Chien
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - Jason L Guo
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - Brandon T Smith
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - Emma Watson
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - Hannah A Pearce
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - Gerry L Koons
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - Adam M Navara
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - Johnny Lam
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - David W Scott
- Department of Statistics, Rice University, 6100 Main Street, Houston, TX 77005, United States of America
| | - K Jane Grande-Allen
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, United States of America.
| |
Collapse
|
45
|
Dai W, Sun M, Leng X, Hu X, Ao Y. Recent Progress in 3D Printing of Elastic and High-Strength Hydrogels for the Treatment of Osteochondral and Cartilage Diseases. Front Bioeng Biotechnol 2020; 8:604814. [PMID: 33330436 PMCID: PMC7729093 DOI: 10.3389/fbioe.2020.604814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/03/2020] [Indexed: 11/13/2022] Open
Abstract
Despite considerable progress for the regenerative medicine, repair of full-thickness articular cartilage defects and osteochondral interface remains challenging. This low efficiency is largely due to the difficulties in recapitulating the stratified zonal architecture of articular cartilage and engineering complex gradients for bone-soft tissue interface. This has led to increased interest in three-dimensional (3D) printing technologies in the field of musculoskeletal tissue engineering. Printable and biocompatible hydrogels are attractive materials for 3D printing applications because they not only own high tunability and complexity, but also offer favorable biomimetic environments for live cells, such as porous structure, high water content, and bioactive molecule incorporation. However, conventional hydrogels are usually mechanically weak and brittle, which cannot reach the mechanical requirements for repair of articular cartilage defects and osteochondral interface. Therefore, the development of elastic and high-strength hydrogels for 3D printing in the repairment of cartilage defects and osteochondral interface is crucial. In this review, we summarized the recent progress in elastic and high-strength hydrogels for 3D printing and categorized them into six groups, namely ion bonds interactions, nanocomposites integrated in hydrogels, supramolecular guest-host interactions, hydrogen bonds interactions, dynamic covalent bonds interactions, and hydrophobic interactions. These 3D printed elastic and high-strength hydrogels may provide new insights for the treatment of osteochondral and cartilage diseases.
Collapse
Affiliation(s)
- Wenli Dai
- Beijing Key Laboratory of Sports Injuries, Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Muyang Sun
- Beijing Key Laboratory of Sports Injuries, Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Xi Leng
- Medical Imaging Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoqing Hu
- Beijing Key Laboratory of Sports Injuries, Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Yingfang Ao
- Beijing Key Laboratory of Sports Injuries, Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
46
|
Bonifacio MA, Cochis A, Cometa S, Gentile P, Scalzone A, Scalia AC, Rimondini L, De Giglio E. From the sea to the bee: Gellan gum-honey-diatom composite to deliver resveratrol for cartilage regeneration under oxidative stress conditions. Carbohydr Polym 2020; 245:116410. [DOI: 10.1016/j.carbpol.2020.116410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/03/2020] [Accepted: 05/03/2020] [Indexed: 01/22/2023]
|
47
|
Schmidt S, Abinzano F, Mensinga A, Teßmar J, Groll J, Malda J, Levato R, Blunk T. Differential Production of Cartilage ECM in 3D Agarose Constructs by Equine Articular Cartilage Progenitor Cells and Mesenchymal Stromal Cells. Int J Mol Sci 2020; 21:ijms21197071. [PMID: 32992847 PMCID: PMC7582568 DOI: 10.3390/ijms21197071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Identification of articular cartilage progenitor cells (ACPCs) has opened up new opportunities for cartilage repair. These cells may be used as alternatives for or in combination with mesenchymal stromal cells (MSCs) in cartilage engineering. However, their potential needs to be further investigated, since only a few studies have compared ACPCs and MSCs when cultured in hydrogels. Therefore, in this study, we compared chondrogenic differentiation of equine ACPCs and MSCs in agarose constructs as monocultures and as zonally layered co-cultures under both normoxic and hypoxic conditions. ACPCs and MSCs exhibited distinctly differential production of the cartilaginous extracellular matrix (ECM). For ACPC constructs, markedly higher glycosaminoglycan (GAG) contents were determined by histological and quantitative biochemical evaluation, both in normoxia and hypoxia. Differential GAG production was also reflected in layered co-culture constructs. For both cell types, similar staining for type II collagen was detected. However, distinctly weaker staining for undesired type I collagen was observed in the ACPC constructs. For ACPCs, only very low alkaline phosphatase (ALP) activity, a marker of terminal differentiation, was determined, in stark contrast to what was found for MSCs. This study underscores the potential of ACPCs as a promising cell source for cartilage engineering.
Collapse
Affiliation(s)
- Stefanie Schmidt
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany;
| | - Florencia Abinzano
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
| | - Anneloes Mensinga
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
| | - Jörg Teßmar
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany; (J.T.); (J.G.)
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany; (J.T.); (J.G.)
| | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - Riccardo Levato
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
- Correspondence: (R.L.); (T.B.)
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany;
- Correspondence: (R.L.); (T.B.)
| |
Collapse
|
48
|
Critchley S, Sheehy EJ, Cunniffe G, Diaz-Payno P, Carroll SF, Jeon O, Alsberg E, Brama PAJ, Kelly DJ. 3D printing of fibre-reinforced cartilaginous templates for the regeneration of osteochondral defects. Acta Biomater 2020; 113:130-143. [PMID: 32505800 DOI: 10.1016/j.actbio.2020.05.040] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022]
Abstract
Successful osteochondral defect repair requires regenerating the subchondral bone whilst simultaneously promoting the development of an overlying layer of articular cartilage that is resistant to vascularization and endochondral ossification. During skeletal development articular cartilage also functions as a surface growth plate, which postnatally is replaced by a more spatially complex bone-cartilage interface. Motivated by this developmental process, the hypothesis of this study is that bi-phasic, fibre-reinforced cartilaginous templates can regenerate both the articular cartilage and subchondral bone within osteochondral defects created in caprine joints. To engineer mechanically competent implants, we first compared a range of 3D printed fibre networks (PCL, PLA and PLGA) for their capacity to mechanically reinforce alginate hydrogels whilst simultaneously supporting mesenchymal stem cell (MSC) chondrogenesis in vitro. These mechanically reinforced, MSC-laden alginate hydrogels were then used to engineer the endochondral bone forming phase of bi-phasic osteochondral constructs, with the overlying chondral phase consisting of cartilage tissue engineered using a co-culture of infrapatellar fat pad derived stem/stromal cells (FPSCs) and chondrocytes. Following chondrogenic priming and subcutaneous implantation in nude mice, these bi-phasic cartilaginous constructs were found to support the development of vascularised endochondral bone overlaid by phenotypically stable cartilage. These fibre-reinforced, bi-phasic cartilaginous templates were then evaluated in clinically relevant, large animal (caprine) model of osteochondral defect repair. Although the quality of repair was variable from animal-to-animal, in general more hyaline-like cartilage repair was observed after 6 months in animals treated with bi-phasic constructs compared to animals treated with commercial control scaffolds. This variability in the quality of repair points to the need for further improvements in the design of 3D bioprinted implants for joint regeneration. STATEMENT OF SIGNIFICANCE: Successful osteochondral defect repair requires regenerating the subchondral bone whilst simultaneously promoting the development of an overlying layer of articular cartilage. In this study, we hypothesised that bi-phasic, fibre-reinforced cartilaginous templates could be leveraged to regenerate both the articular cartilage and subchondral bone within osteochondral defects. To this end we used 3D printed fibre networks to mechanically reinforce engineered transient cartilage, which also contained an overlying layer of phenotypically stable cartilage engineered using a co-culture of chondrocytes and stem cells. When chondrogenically primed and implanted into caprine osteochondral defects, these fibre-reinforced bi-phasic cartilaginous grafts were shown to spatially direct tissue development during joint repair. Such developmentally inspired tissue engineering strategies, enabled by advances in biofabrication and 3D printing, could form the basis of new classes of regenerative implants in orthopaedic medicine.
Collapse
Affiliation(s)
- Susan Critchley
- 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
| | - Eamon J Sheehy
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre, Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Gráinne 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
| | - Pedro Diaz-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
| | - Simon F Carroll
- 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
| | - Oju Jeon
- Department of Bioengineering, University of Illinois, Chicago, IL, USA
| | - Eben Alsberg
- Department of Bioengineering, University of Illinois, Chicago, IL, USA; Departments of Orthopaedics, Pharmacology, and Mechanical & Industrial Engineering, University of Illinois, Chicago, IL, USA
| | - Pieter A J Brama
- School of Veterinary Medicine, University 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, Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
49
|
Yao Y, Zhang T, Chen H, Zheng S, Chen Y, Zhang S. Enhanced chondrogenesis in a coculture system with genetically manipulated dedifferentiated chondrocytes and ATDC5 cells. Biotechnol Bioeng 2020; 117:3173-3181. [PMID: 32633811 DOI: 10.1002/bit.27482] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/17/2020] [Accepted: 07/05/2020] [Indexed: 12/12/2022]
Abstract
Articular cartilage repair after injury is a great challenge worldwide due to its nerveless and avascular features. Tissue engineering is proposed as a promising alternative for cartilage regeneration. In this study, an adenoviral vector carrying the transforming growth factor-β3 (TGF-β3) gene was constructed and introduced into dedifferentiated chondrocytes, which were then cocultured with ATDC5 cells in an alginate hydrogel system. The results showed that the experimental groups exhibited better cell viability and higher levels of cartilage-related genes than the control groups. In this coculture system, the chondrogenic differentiation of ATDC5 cells was effectively induced by TGF-β3 and other latent cytokines that were produced by the transfected chondrocytes. Thus, this method can avoid the degradation of exogenous TGF-β3, and it can protect ATDC5 cells during virus transfection to maintain cell viability and chondrogenic differentiation capability. Taken together, this study provides fresh insights for applying this genetically manipulated coculture system to cartilage repair in the future.
Collapse
Affiliation(s)
- Yongchang Yao
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou, China
| | - Tingshuai Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Orthopaedics, Dongguan Tungwah Hospital, Dongguan, China
| | - Hanzheng Chen
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou, China
| | - Shicong Zheng
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou, China
| | - Yi Chen
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou, China
| | - Shujiang Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou, China
| |
Collapse
|
50
|
Chen J, Chin A, Almarza AJ, Taboas JM. Hydrogel to guide chondrogenesis versus osteogenesis of mesenchymal stem cells for fabrication of cartilaginous tissues. Biomed Mater 2020; 15:045006. [PMID: 31470441 PMCID: PMC11934051 DOI: 10.1088/1748-605x/ab401f] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The ideal combination of hydrogel components for regeneration of cartilage and cartilaginous interfaces is a significant challenge because control over differentiation into multiple lineages is necessary. Stabilization of the phenotype of stem cell derived chondrocytes is needed to avoid undesired progression to terminal hypertrophy and tissue mineralization. A novel ternary blend hydrogel composed of methacrylated poly(ethylene glycol) (PEG), gelatin, and heparin (PGH) was designed to guide chondrogenesis by bone marrow derived mesenchymal stem cells (BMSCs) and maintenance of their cartilaginous phenotype. The hydrogel material effects on chondrogenic and osteogenic differentiation by BMSCs were evaluated in comparison to methacrylated gelatin hydrogel (GEL), a conventional bioink used for both chondrogenic and osteogenic applications. PGH and GEL hydrogels were loaded with goat BMSCs and cultured in chondrogenic and osteogenic mediums in vitro over six weeks. The PGH showed no sign of mineral deposition in an osteogenic environment in vitro. To further evaluate material effects, the hydrogels were loaded with adult human BMSCs (hBMSCs) and transforming growth factor β-3 and grown in subcutaneous pockets in mice over eight weeks. Consistent with the in vitro results, the PGH had greater potential to induce chondrogenesis by BMSCs in vivo compared to the GEL as evidenced by elevated gene expression of chondrogenic markers, supporting its potential for stable cartilage engineering. The PGH also showed a greater percentage of GAG positive cells compared to the GEL. Unlike the GEL, the PGH hydrogel exhibited anti-osteogenic effects in vivo as evidenced by negative Von Kossa staining and suppressed gene expression of hypertrophic and osteogenic markers. By nature of their polymer composition alone, the PGH and GEL regulated BMSC differentiation down different osteochondral lineages. Thus, the PGH and GEL are promising hydrogels to regenerate stratified cartilaginous interfacial tissues in situ, such as the mandibular condyle surface, using undifferentiated BMSCs and a stratified scaffold design.
Collapse
Affiliation(s)
- Jingming Chen
- Department of Bioengineering; University of Pittsburgh, Pittsburgh, PA 15213, United States of America. Center for Craniofacial Regeneration; University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | | | | | | |
Collapse
|