1
|
Qian Y, Gu Y, Tribukait-Riemenschneider F, Martin I, Shastri VP. Incorporation of Cross-Linked Gelatin Microparticles To Enhance Cell Attachment and Chondrogenesis in Carboxylated Agarose Bioinks for Cartilage Engineering. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22293-22307. [PMID: 40194271 PMCID: PMC12012782 DOI: 10.1021/acsami.5c00077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/09/2025]
Abstract
Due to the limited regenerative capacity of injured cartilage, surgical intervention using engineered cellular constructs or autologous cell implantation is the best accredited approach to prevent further degeneration and promote a regenerative microenvironment. Advancements in additive manufacturing present opportunities for graft customization through enhanced scaffold design. In bioprinting, an additive manufacturing process, the "bioink" serves as the medium to carry cells but also as a scaffold by imparting form and mechanical attributes to the printed object. In this study, the impact of cross-linked gelatin microparticles (GMPs) on rheological properties and printability of carboxylated agarose (CA) bioink as well as matrix deposition by human nasal chondrocytes (hNCs) was investigated. The introduction of GMPs yielded stiffer bioink formulations, with lower sol-gel transitions that retained the exceptional printability of CA. GMPs served as foci for the attachment of hNCs, improving cellular distribution and bridging the deposited extracellular matrix. After 4 weeks in chondrogenic culture, GMPs containing printed constructs showed enhanced toughness approaching that of the lower end of the spectrum of native cartilage tissue. The incorporation of proteinaceous microparticles might serve as a general concept to promote cellular function in polysaccharide-based bioinks and opens another avenue for engineering 3D-bioprinted microenvironments.
Collapse
Affiliation(s)
- Yi Qian
- Institute
for Macromolecular Chemistry, University
of Freiburg, Freiburg 79104, Germany
| | - Yawei Gu
- Institute
for Macromolecular Chemistry, University
of Freiburg, Freiburg 79104, Germany
| | | | - Ivan Martin
- Department
of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - V. Prasad Shastri
- Institute
for Macromolecular Chemistry, University
of Freiburg, Freiburg 79104, Germany
- BIOSS—Centre
for Biological Signalling Studies, University
of Freiburg, Freiburg 79104, Germany
| |
Collapse
|
2
|
Kutaish H, Klopfenstein A, Obeid Adorisio SN, Tscholl PM, Fucentese S. Current trends in the treatment of focal cartilage lesions: a comprehensive review. EFORT Open Rev 2025; 10:203-212. [PMID: 40167465 PMCID: PMC12002736 DOI: 10.1530/eor-2024-0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Focal cartilage lesions refer to localized damage or defects in the cartilage covering joint surfaces, often resulting from trauma, wear and tear or underlying joint conditions. These lesions can lead to pain, impaired joint function and, if left untreated, may contribute to the development of degenerative joint diseases. Challenges in treatment of focal cartilage lesion are mainly due to limited intrinsic healing capacity, difficulty in early detection of lesions and variability in symptoms make timely intervention tricky. Conservative treatments varies from addressing symptoms using physical therapy, corticosteroid injections and viscosupplementation, to regenerative attempts such as in platelet-rich plasma and mesenchymal stem cells therapy. These modalities provide a limited duration of improvement and are commonly used to delay more aggressive treatment. Traditional surgery options are mainly summed up by microfractures (MFX) for smaller lesions, osteochondral autograft transfer, osteochondral allograft transfer (OCA) and autologous matrix-induced chondrogenesis for moderate-to-large lesions. Cellular approaches encompass autologous chondrocyte implantation (ACI), which involve targeted transplantation of chondrocytes. Current research is concentrating on cell-based surgical approaches utilizing advanced biomaterials for both scaffold and scaffold-free implants. While gene therapy and tissue engineering approaches aim to optimize chondrocyte proliferation and differentiation for improved quality of the transplanted biomaterial and patient's outcomes.
Collapse
|
3
|
Decoene I, Svitina H, Hamed MB, Economou A, Stegen S, Luyten FP, Papantoniou I. Callus organoids reveal distinct cartilage to bone transition mechanisms across donors and a role for biological sex. Bone Res 2025; 13:41. [PMID: 40140357 PMCID: PMC11947321 DOI: 10.1038/s41413-025-00418-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Clinical translation of tissue-engineered advanced therapeutic medicinal products is hindered by a lack of patient-dependent and independent in-process biological quality controls that are reflective of in vivo outcomes. Recent insights into the mechanism of native bone repair highlight a robust path dependence. Organoid-based bottom-up developmental engineering mimics this path-dependence to design personalized living implants scaffold-free, with in-build outcome predictability. Yet, adequate (noninvasive) quality metrics of engineered tissues are lacking. Moreover, insufficient insight into the role of donor variability and biological sex as influencing factors for the mechanism toward bone repair hinders the implementation of such protocols for personalized bone implants. Here, male and female bone-forming organoids were compared to non-bone-forming organoids regarding their extracellular matrix composition, transcriptome, and secreted proteome signatures to directly link in vivo outcomes to quality metrics. As a result, donor variability in bone-forming callus organoids pointed towards two distinct pathways to bone, through either a hypertrophic cartilage or a fibrocartilaginous template. The followed pathway was determined early, as a biological sex-dependent activation of distinct progenitor populations. Independent of donor or biological sex, a cartilage-to-bone transition was driven by a common panel of secreted factors that played a role in extracellular matrix remodeling, mineralization, and attraction of vasculature. Hence, the secreted proteome is a source of noninvasive biomarkers that report on biological potency and could be the missing link toward data-driven decision-making in organoid-based bone tissue engineering.
Collapse
Affiliation(s)
- Isaak Decoene
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
| | - Hanna Svitina
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
| | - Mohamed Belal Hamed
- Laboratory of Molecular Bacteriology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Molecular Biology Department, National Research Centre, 33 El Buhouth st, Dokii, 12622, Cairo, Egypt
- Department of Neurosciences, Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven, VIB-KU Leuven Center for Brain & Disease Research, Herestraat, 3000, Leuven, Belgium
| | - Anastassios Economou
- Laboratory of Molecular Bacteriology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Steve Stegen
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Frank P Luyten
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
| | - Ioannis Papantoniou
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium.
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium.
- Institute for Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH), Stadiou Street, Platani, box 1414, 26504, Patras, Greece.
| |
Collapse
|
4
|
Pico OA, Espinoza F, Cádiz MI, Sossa CL, Becerra-Bayona SM, Salgado MCC, Rodríguez JER, Cárdenas OFV, Cure JMQ, Khoury M, Arango-Rodríguez ML. Efficacy of a single dose of cryopreserved human umbilical cord mesenchymal stromal cells for the treatment of knee osteoarthritis:a randomized, controlled, double-blind pilot study. Cytotherapy 2025; 27:188-200. [PMID: 39503681 DOI: 10.1016/j.jcyt.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/14/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Knee osteoarthritis (OA) is the most prevalent degenerative musculoskeletal disorder, which is particularly common in older population. While conventional treatments have limited effectiveness, the development of more effective therapeutic strategies is necessary to address this primary source of pain and disability. Umbilical cord mesenchymal stromal cells (UC-MSCs) offer a promising therapeutic approach for treating knee OA. AIM This randomized, prospective, double-blind and controlled pilot study was carried out to evaluate and compare the safety and therapeutic efficacy of a single intra-articular injection of a standardized product CellistemOA (5 × 106 ± 5 × 105 UC-MSCs), vs. triamcinolone (a synthetic corticosteroid) (10 mg/mL) in thirty patients with symptomatic knee OA (Kellgren-Lawrence grade II or III). METHODS The outcomes included changes in Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores based on a Likert scale, numerical rating score (NRS) for pain, Magnetic Resonance Imaging (MRI), and quality of life (SF-36 questionnaire), from baseline and throughout 12-months of follow-up. RESULTS Patients treated with CellistemOA showed significant improvement in WOMAC score (including the three subscale scores (pain, stiffness and function), NRS in pain, and SF-36 profile from baseline to 12 months (p < 0.05) compared to the triamcinolone group, and no severe adverse events were reported. There were no significant differences in MRI WORMS scores between the two groups. However, patients who received the cellular treatment experienced a significant improvement in their SF-36 profile (p < 0.05). CONCLUSIONS This pilot study revealed that a single dose of CellistemOA is safe and superior to the active comparator in knee OA at 1-year of follow-up, making it a compelling therapeutic alternative to treat symptomatic OA patients.
Collapse
Affiliation(s)
- Omar Amado Pico
- Fundación Oftalmológica de Santander - FOSCAL, Floridablanca, Colombia; Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | - Francisco Espinoza
- Cells for Cells & Consorcio Regenero, Santiago, Chile; Program for Translational Research in Cell Therapy, Universidad de los Andes, Santiago, Chile; Department of Rheumatology, Universidad de los Andes, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María Ignacia Cádiz
- Cells for Cells & Consorcio Regenero, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Laboratory of Nano-Regenerative Medicine, Universidad de los Andes, Santiago, Chile
| | - Claudia L Sossa
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia; Programa para el Tratamiento y Estudio de Enfermedades Hematológicas y Oncológicas de Santander (PROTEHOS), Floridablanca, Colombia
| | - Silvia M Becerra-Bayona
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | - María C Canencio Salgado
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | | | | | | | - Maroun Khoury
- Cells for Cells & Consorcio Regenero, Santiago, Chile; Program for Translational Research in Cell Therapy, Universidad de los Andes, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Laboratory of Nano-Regenerative Medicine, Universidad de los Andes, Santiago, Chile
| | - Martha L Arango-Rodríguez
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia; Banco Multitejidos y Centro de Terapias Avanzadas, Clínica FOSCAL Internacional, Floridablanca, Colombia.
| |
Collapse
|
5
|
Heo S, Noh M, Kim Y, Park S. Stem Cell-Laden Engineered Patch: Advances and Applications in Tissue Regeneration. ACS APPLIED BIO MATERIALS 2025; 8:62-87. [PMID: 39701826 DOI: 10.1021/acsabm.4c01427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Stem cell-based therapies are emerging as significant approaches in tissue engineering and regenerative medicine, applicable to both fundamental scientific research and clinical practice. Despite remarkable results in clinical studies, challenges such as poor standardization of graft tissues, limited sources, and reduced functionality have hindered the effectiveness of these therapies. In this review, we summarize the engineering approaches involved in fabricating stem cell assisted patches and the substantial strategies for designing stem cell-laden engineered patches (SCP) to complement the existing stem cell-based therapies. We then outline the potential applications of SCP in advancing tissue regeneration and regenerative medicine. By combining living stem cells with engineered patches, SCP can enhance the functions of both components, particularly for tissue engineering applications. Finally, we addressed current challenges, such as ethical considerations, high costs, and regulatory hurdles and proposed future research directions to overcome these barriers.
Collapse
Affiliation(s)
- Seyeong Heo
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Minhyeok Noh
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Yeonseo Kim
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Sunho Park
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| |
Collapse
|
6
|
Mathur V, Dsouza V, Srinivasan V, Vasanthan KS. Volumetric Additive Manufacturing for Cell Printing: Bridging Industry Adaptation and Regulatory Frontiers. ACS Biomater Sci Eng 2025; 11:156-181. [PMID: 39746181 PMCID: PMC11733917 DOI: 10.1021/acsbiomaterials.4c01837] [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: 10/03/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
Volumetric additive manufacturing (VAM) is revolutionizing the field of cell printing by enabling the rapid creation of complex three-dimensional cellular structures that mimic natural tissues. This paper explores the advantages and limitations of various VAM techniques, such as holographic lithography, digital light processing, and volumetric projection, while addressing their suitability across diverse industrial applications. Despite the significant potential of VAM, challenges related to regulatory compliance and scalability persist, particularly in the context of bioprinted tissues. In India, the lack of clear regulatory guidelines and intellectual property protections poses additional hurdles for companies seeking to navigate the evolving landscape of bioprinting. This study emphasizes the importance of collaboration among industry stakeholders, regulatory agencies, and academic institutions to establish tailored frameworks that promote innovation while ensuring safety and efficacy. By bridging the gap between technological advancement and regulatory oversight, VAM can unlock new opportunities in regenerative medicine and tissue engineering, transforming patient care and therapeutic outcomes.
Collapse
Affiliation(s)
- Vidhi Mathur
- Manipal
Centre for Biotherapeutics Research, Manipal
Academy of Higher Education, Manipal, 576104 Karnataka, India
| | - Vinita Dsouza
- Manipal
Centre for Biotherapeutics Research, Manipal
Academy of Higher Education, Manipal, 576104 Karnataka, India
| | - Varadharajan Srinivasan
- Department
of Civil Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104 Karnataka, India
| | - Kirthanashri S Vasanthan
- Manipal
Centre for Biotherapeutics Research, Manipal
Academy of Higher Education, Manipal, 576104 Karnataka, India
| |
Collapse
|
7
|
Torabi Rahvar P, Abdekhodaie MJ, Jooybar E, Gantenbein B. An enzymatically crosslinked collagen type II/hyaluronic acid hybrid hydrogel: A biomimetic cell delivery system for cartilage tissue engineering. Int J Biol Macromol 2024; 279:134614. [PMID: 39127277 DOI: 10.1016/j.ijbiomac.2024.134614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
This study presents new injectable hydrogels based on hyaluronic acid and collagen type II that mimic the polysaccharide-protein structure of natural cartilage. After collagen isolation from chicken sternal cartilage, tyramine-grafted hyaluronic acid and collagen type II (HA-Tyr and COL-II-Tyr) were synthesized. Hybrid hydrogels were prepared with different ratios of HA-Tyr/COL-II-Tyr using horseradish peroxidase and noncytotoxic concentrations of hydrogen peroxide to encapsulate human bone marrow-derived mesenchymal stromal cells (hBM-MSCs). The findings showed that a higher HA-Tyr content resulted in a higher storage modulus and a lower hydrogel shrinkage, resulting in hydrogel swelling. Incorporating COL-II-Tyr into HA-Tyr hydrogels induced a more favorable microenvironment for hBM-MSCs chondrogenic differentiation. Compared to HA-Tyr alone, the hybrid HA-Tyr/COL-II-Tyr hydrogel promoted enhanced chondrocyte adhesion, spreading, proliferation, and upregulation of cartilage-related gene expression. These results highlight the promising potential of injectable HA-Tyr/COL-II-Tyr hybrid hydrogels to deliver cells for cartilage regeneration.
Collapse
Affiliation(s)
- Parisa Torabi Rahvar
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | - Mohammad J Abdekhodaie
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Environmental and Applied Science Management, Yeates School of Graduate Studies, Toronto Metropolitan University, Toronto, Canada.
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland; Inselspital, Bern University Hospital, Department of Orthopedic Surgery & Traumatology, Bern, Switzerland
| |
Collapse
|
8
|
Court AC, Vega-Letter AM, Parra-Crisóstomo E, Velarde F, García C, Ortloff A, Vernal R, Pradenas C, Luz-Crawford P, Khoury M, Figueroa FE. Mitochondrial transfer balances cell redox, energy and metabolic homeostasis in the osteoarthritic chondrocyte preserving cartilage integrity. Theranostics 2024; 14:6471-6486. [PMID: 39479450 PMCID: PMC11519804 DOI: 10.7150/thno.96723] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/22/2024] [Indexed: 11/02/2024] Open
Abstract
Osteoarthrosis (OA) is a leading cause of disability and early mortality, with no disease modifying treatment. Mitochondrial (MT) dysfunction and changes in energy metabolism, leading to oxidative stress and apoptosis, are main drivers of disease. In reaction to stress, mesenchymal stromal/stem cells (MSCs) donate their MT to damaged tissues. Methods: To evaluate the capacity of clinically validated MSCs to spontaneously transfer their MT to human OA chondrocytes (OA-Ch), primary cultured Ch isolated from the articular cartilage of OA patients were co-cultured with MT-labeled MSCs. MT transfer (MitoT) was evidenced by flow cytometry and confocal microscopy of MitoTracker-stained and YFP-tagged MT protein. MT persistence and metabolic analysis on target cells were assessed by direct transfer of MSC-derived MT to OA-Chs (Mitoception), through SNP-qPCR analysis, ATP measurements and Seahorse technology. The effects of MitoT on MT dynamics, oxidative stress and cell viability were gauged by western blot of fusion/fission proteins, confocal image analysis, ROS levels, Annexin V/7AAD and TUNEL assays. Intra-articular injection of MSC-derived MT was tested in a collagenase-induced murine model of OA. Results: Dose-dependent cell-to-cell MitoT from MSCs to cultured OA-Chs was detected starting at 4 hours of co-culture, with increasing MT-fluorescence levels at higher MSC:Ch ratios. PCR analysis confirmed the presence of exogenous MSC-MT within MitoT+ OA-Chs up to 9 days post Mitoception. MitoT from MSCs to OA-Ch restores energetic status, with a higher ATP production and metabolic OXPHOS/Glycolisis ratio. Significant changes in the expression of MT network regulators, increased MFN2 and decreased p-DRP1, reveal that MitoT promotes MT fusion restoring the MT dynamics in the OA-Ch. Additionally, MitoT increases SOD2 transcripts, protein, and activity levels, and reduces ROS levels, confering resistance to oxidative stress and enhancing resistance to apoptosis. Intra-articular injection of MSC-derived MT improves histologic scores and bone density of the affected joints in the OA mouse model, demonstrating a protective effect of MT transplantation on cartilage degradation. Conclusion: The Mitochondria transfer of MSC-derived MT induced reversal of the metabolic dysfunction by restoring the energetic status and mitochondrial dynamics in the OA chondrocyte, while conferring resistance to oxidative stress and apoptosis. Intra-articular injection of MT improved the disease in collagenase-induced OA mouse model. The restoration of the cellular homeostasis and the preclinical benefit of the intra-articular MT treatment offer a new approach for the treatment of OA.
Collapse
Affiliation(s)
- Angela C. Court
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ana María Vega-Letter
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliseo Parra-Crisóstomo
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Francesca Velarde
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Rolando Vernal
- Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Carolina Pradenas
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Patricia Luz-Crawford
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Maroun Khoury
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Fernando E. Figueroa
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
9
|
Parasuraman G, Amirtham SM, Francis DV, Livingston A, Ramasamy B, Sathishkumar S, Vinod E. Evaluation of Chondral Defect Repair Using Human Fibronectin Adhesion Assay-Derived Chondroprogenitors Suspended in Lyophilized Fetal Collagen Scaffold: An Ex Vivo Osteochondral Unit Model Study. Indian J Orthop 2024; 58:991-1000. [PMID: 39087036 PMCID: PMC11286923 DOI: 10.1007/s43465-024-01192-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/22/2024] [Indexed: 08/02/2024]
Abstract
Introduction Chondral defect repair is challenging due to a scarcity of reparative cells and the need to fill a large surface area, compounded by the absence of self-healing mechanisms. Fibronectin adhesion assay-derived chondroprogenitors (FAA-CPs) have emerged as a promising alternative with enhanced chondrogenic ability and reduced hypertrophy. De-cellularized bio-scaffolds are reported to act as extracellular matrix, mimicking the structural and functional characteristics of native tissue, thereby facilitating cell attachment and differentiation. This study primarily assessed the synergistic effect of FAA-CPs suspended in fetal cartilage-derived collagen-containing scaffolds in repairing chondral defects. Methodology The de-cellularized and lyophilized fetal collagen was prepared from the tibio-femoral joint of a 36 + 4-week gestational age fetus. FAA-CPs were isolated from osteoarthritic cartilage samples (n = 3) and characterized. In ex vivo analysis, FAA-CPs at a density of 1 × 106 cells were suspended in the lyophilized scaffold and placed into the chondral defects created in the Osteochondral Units and harvested on the 35th day for histological examination. Results The lyophilized scaffold of de-cellularized fetal cartilage with FAA-CPs demonstrated effective healing of the critical size chondral defect. This was evidenced by a uniform distribution of cells, a well-organized collagen-fibrillar network, complete filling of the defect with alignment to the surface, and favorable integration with the adjacent cartilage. However, these effects were less pronounced in the plain scaffold control group and no demonstrable repair observed in the empty defect group. Conclusion This study suggests the synergistic potential of FAA-CPs and collagen scaffold for chondral repair which needs to be further explored for clinical therapy. Supplementary Information The online version contains supplementary material available at 10.1007/s43465-024-01192-6.
Collapse
Affiliation(s)
- Ganesh Parasuraman
- Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | - Soosai Manickam Amirtham
- Department of Physiology/Centre for Stem Cell Research, Christian Medical College, Vellore, India
| | | | - Abel Livingston
- Department of Orthopaedics, Christian Medical College, Vellore, India
| | - Boopalan Ramasamy
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
- Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, Australia
| | - Solomon Sathishkumar
- Department of Physiology/Centre for Stem Cell Research, Christian Medical College, Vellore, India
| | - Elizabeth Vinod
- Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India
- Department of Physiology/Centre for Stem Cell Research, Christian Medical College, Vellore, India
| |
Collapse
|
10
|
Sharma D, Satapathy BK. Nanostructured Biopolymer-Based Constructs for Cartilage Regeneration: Fabrication Techniques and Perspectives. Macromol Biosci 2024; 24:e2400125. [PMID: 38747219 DOI: 10.1002/mabi.202400125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/08/2024] [Indexed: 05/24/2024]
Abstract
The essential functions of cartilage, such as shock absorption and resilience, are hindered by its limited regenerative capacity. Although current therapies alleviate symptoms, novel strategies for cartilage regeneration are desperately needed. Recent developments in three-dimensional (3D) constructs aim to address this challenge by mimicking the intrinsic characteristics of native cartilage using biocompatible materials, with a significant emphasis on both functionality and stability. Through fabrication methods such as 3D printing and electrospinning, researchers are making progress in cartilage regeneration; nevertheless, it is still very difficult to translate these advances into clinical practice. The review emphasizes the importance of integrating various fabrication techniques to create stable 3D constructs. Meticulous design and material selection are required to achieve seamless cartilage integration and durability. The review outlines the need to address these challenges and focuses on the latest developments in the production of hybrid 3D constructs based on biodegradable and biocompatible polymers. Furthermore, the review acknowledges the limitations of current research and provides perspectives on potential avenues for effectively regenerating cartilage defects in the future.
Collapse
Affiliation(s)
- Deepika Sharma
- Department of Materials Science and Engineering, Indian Institute of Technology Delhi, Delhi, India
- Department of Food Science, The Pennsylvania State University, University Park, PA, USA
| | - Bhabani K Satapathy
- Department of Materials Science and Engineering, Indian Institute of Technology Delhi, Delhi, India
| |
Collapse
|
11
|
Kotlier JL, Yazditabar JM, Fathi A, Mayfield CK, Ahmad A, Petrigliano FA, Liu JN. Industry affiliation does not predict outcomes of randomized controlled trials for mesenchymal stem cells in knee osteoarthritis. Knee 2024; 49:1-7. [PMID: 38824767 DOI: 10.1016/j.knee.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/31/2024] [Accepted: 05/09/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND This research aims to determine the influence of industry on the outcomes of randomized controlled trials (RCTs) for Mesenchymal Stem Cell (MSC) treatments in knee osteoarthritis (OA). METHODS PubMed, Scopus, and Web of Science were searched from 2010 onwards using the terms "knee osteoarthritis" and "mesenchymal stem cells". After identifying relevant RCTs, studies were categorized as industry-affiliated or non-industry-affiliated. They were also classified as favorable if they achieved statistically significant (p < 0.05) results with MSC injections compared to control. Chi-squared tests were employed to analyze the relationship between industry affiliation and study outcome. RESULTS Post exclusion criteria, 38 studies were analyzed. Of these, there were 20 (52.6%) industry affiliated (IA) and 18 (47.4%) non-industry affiliated (NIA) studies. Among the 20 IA studies, 17 (85.0%) reported favorable outcomes for MSC treatment arm, with the remaining 3 (15.0%) showing analogous (no difference between treatment arms) results. For the 18 NIA studies, 15 (83.3%) were favorable, and 3 (16.6%) were analogous. No significant difference in outcomes was observed between IA and NIA studies (p = 0.888). Analysis of patient reported outcomes also revealed no significant difference. Of note, studies using allogeneic MSCs were more likely to be IA than studies using autologous MSCs (p = 0.005) CONCLUSION: This study demonstrated no strong association between industry affiliation and the outcomes of RCTs for MSC treatments in knee OA. Despite this, the potential influence of industry ties should always be considered when applying study findings to new treatment modalities for patient care.
Collapse
Affiliation(s)
| | | | - Amir Fathi
- USC Keck School of Medicine, Los Angeles, CA, USA
| | | | - Aamir Ahmad
- USC Keck School of Medicine, Los Angeles, CA, USA
| | | | - Joseph N Liu
- USC Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
12
|
Lele M, Kapur S, Hargett S, Sureshbabu NM, Gaharwar AK. Global trends in clinical trials involving engineered biomaterials. SCIENCE ADVANCES 2024; 10:eabq0997. [PMID: 39018412 PMCID: PMC466960 DOI: 10.1126/sciadv.abq0997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/11/2024] [Indexed: 07/19/2024]
Abstract
Engineered biomaterials are materials specifically designed to interact with biological systems for biomedical applications. This paper offers the comprehensive analysis of global clinical trial trends involving such materials. We surveyed 834 studies in the ClinicalTrials.gov database and explored biomaterial types, their initiation points, and durations in clinical trials. Predominantly, synthetic and natural polymers, particularly silicone and collagen, are used. Trials, focusing on ophthalmology, dentistry, and vascular medicine, are primarily conducted in the United States, Canada, and Italy. These trials encompass a broad demographic, and trials enrolled fewer than 100 participants. The study duration varied ranging from 0.5 to 4.5 years. These biomaterials are mainly bioresorbable or bioinert, with the integration of cells in biomaterials remaining an underexplored area. Our findings shed light on current practices and future potentials of engineered biomaterials in clinical research, offering insights for advancing this dynamic field globally.
Collapse
Affiliation(s)
- Mahim Lele
- Bridgeland High School, 10707 Mason Rd., Cypress, TX 77433, USA
| | - Shaunak Kapur
- Seven Lakes High School, 9251 S Fry Rd., Katy, TX 77494, USA
| | - Sarah Hargett
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Nivedhitha Malli Sureshbabu
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA
- Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX 77843, USA
- Department of Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
13
|
Zhou H, Zhang Z, Mu Y, Yao H, Zhang Y, Wang DA. Harnessing Nanomedicine for Cartilage Repair: Design Considerations and Recent Advances in Biomaterials. ACS NANO 2024; 18:10667-10687. [PMID: 38592060 DOI: 10.1021/acsnano.4c00780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Cartilage injuries are escalating worldwide, particularly in aging society. Given its limited self-healing ability, the repair and regeneration of damaged articular cartilage remain formidable challenges. To address this issue, nanomaterials are leveraged to achieve desirable repair outcomes by enhancing mechanical properties, optimizing drug loading and bioavailability, enabling site-specific and targeted delivery, and orchestrating cell activities at the nanoscale. This review presents a comprehensive survey of recent research in nanomedicine for cartilage repair, with a primary focus on biomaterial design considerations and recent advances. The review commences with an introductory overview of the intricate cartilage microenvironment and further delves into key biomaterial design parameters crucial for treating cartilage damage, including microstructure, surface charge, and active targeting. The focal point of this review lies in recent advances in nano drug delivery systems and nanotechnology-enabled 3D matrices for cartilage repair. We discuss the compositions and properties of these nanomaterials and elucidate how these materials impact the regeneration of damaged cartilage. This review underscores the pivotal role of nanotechnology in improving the efficacy of biomaterials utilized for the treatment of cartilage damage.
Collapse
Affiliation(s)
- Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Yi Zhang
- School of Integrated Circuit Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
- Center for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
14
|
Chu H, Zhang S, Zhang Z, Yue H, Liu H, Li B, Yin F. Comparison studies identify mesenchymal stromal cells with potent regenerative activity in osteoarthritis treatment. NPJ Regen Med 2024; 9:14. [PMID: 38561335 PMCID: PMC10984924 DOI: 10.1038/s41536-024-00358-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Osteoarthritis affects 15% of people over 65 years of age. It is characterized by articular cartilage degradation and inflammation, leading to joint pain and disability. Osteoarthritis is incurable and the patients may eventually need joint replacement. An emerging treatment is mesenchymal stromal cells (MSCs), with over two hundred clinical trials being registered. However, the outcomes of these trials have fallen short of the expectation, due to heterogeneity of MSCs and uncertain mechanisms of action. It is generally believed that MSCs exert their function mainly by secreting immunomodulatory and trophic factors. Here we used knee osteoarthritis mouse model to assess the therapeutic effects of MSCs isolated from the white adipose or dermal adipose tissue of Prrx1-Cre; R26tdTomato mice and Dermo1-Cre; R26tdTomato mice. We found that the Prrx1-lineage MSCs from the white adipose tissues showed the greatest in vitro differentiation potentials among the four MSC groups and single cell profiling showed that the Prrx1-lineage MSCs contained more stem cells than the Dermo1 counterpart. Only the Prrx1-lineage cells isolated from white adipose tissues showed long-term therapeutic effectiveness on early-stage osteoarthritis models. Mechanistically, Prrx1-lineage MSCs differentiated into Col2+ chondrocytes and replaced the damage cartilage, activated Col1 expressing in resident chondrocytes, and inhibited synovial inflammation. Transcriptome analysis showed that the articular chondrocytes derived from injected MSCs expressed immunomodulatory cytokines, trophic factors, and chondrocyte-specific genes. Our study identified a MSC population genetically marked by Prrx1 that has great multipotentiality and can differentiate into chondrocytes to replace the damaged cartilage.
Collapse
Affiliation(s)
- Hongshang Chu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shaoyang Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhenlin Zhang
- Department of Osteoporosis and Bone Diseases, Shanghai Clinical Research Center of Bone Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Hua Yue
- Department of Osteoporosis and Bone Diseases, Shanghai Clinical Research Center of Bone Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
| | - Feng Yin
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, 200120, China.
- Department of Joint and Sports Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
15
|
Mehta JM, Hiremath SC, Chilimba C, Ghasemi A, Weaver JD. Translation of cell therapies to treat autoimmune disorders. Adv Drug Deliv Rev 2024; 205:115161. [PMID: 38142739 PMCID: PMC10843859 DOI: 10.1016/j.addr.2023.115161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Autoimmune diseases are a diverse and complex set of chronic disorders with a substantial impact on patient quality of life and a significant global healthcare burden. Current approaches to autoimmune disease treatment comprise broadly acting immunosuppressive drugs that lack disease specificity, possess limited efficacy, and confer undesirable side effects. Additionally, there are limited treatments available to restore organs and tissues damaged during the course of autoimmune disease progression. Cell therapies are an emergent area of therapeutics with the potential to address both autoimmune disease immune dysfunction as well as autoimmune disease-damaged tissue and organ systems. In this review, we discuss the pathogenesis of common autoimmune disorders and the state-of-the-art in cell therapy approaches to (1) regenerate or replace autoimmune disease-damaged tissue and (2) eliminate pathological immune responses in autoimmunity. Finally, we discuss critical considerations for the translation of cell products to the clinic.
Collapse
Affiliation(s)
- Jinal M Mehta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Shivani C Hiremath
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Chishiba Chilimba
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Azin Ghasemi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jessica D Weaver
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
16
|
Peng L, Li H, Deng H, Gao T, Li R, Xu Z, Tian Q, Zhao T, Li J, Yang Y, Wang C, Liu S, Guo Q. Combination of a human articular cartilage-derived extracellular matrix scaffold and microfracture techniques for cartilage regeneration: A proof of concept in a sheep model. J Orthop Translat 2024; 44:72-87. [PMID: 38259590 PMCID: PMC10801125 DOI: 10.1016/j.jot.2023.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/16/2023] [Accepted: 09/20/2023] [Indexed: 01/24/2024] Open
Abstract
Background The utilization of decellularized extracellular matrix has gained considerable attention across numerous areas in regenerative research. Of particular interest is the human articular cartilage-derived extracellular matrix (hACECM), which presents as a promising facilitator for cartilage regeneration. Concurrently, the microfracture (MF) technique, a well-established marrow stimulation method, has proven efficacious in the repair of cartilage defects. However, as of the current literature review, no investigations have explored the potential of a combined application of hACECM and the microfracture technique in the repair of cartilage defects within a sheep model. Hypothesis The combination of hACECM scaffold and microfracture will result in improved repair of full-thickness femoral condyle articular cartilage defects compared to the use of either technique alone. Study design Controlled laboratory study. Methods Full-thickness femoral condyle articular cartilage defect (diameter, 7.0 mm; debrided down to the subchondral bone plate) were created in the weight-bearing area of the femoral medial and lateral condyles (n = 24). All of defected sheep were randomly divided into four groups: control, microfracture, hACECM scaffold, and hACECM scaffold + microfracture. After 3, 6 and 12 months, the chondral repair was assessed for standardized (semi-) quantitative macroscopic, imaging, histological, immunohistochemical, mechanics, and biochemical analyses in each group. Result At 3, 6 and 12 months after implantation, the gross view and pathological staining of regenerative tissues were better in the hACECM scaffold and hACECM scaffold + microfracture groups than in the microfracture and control groups; Micro-CT result showed that the parameters about the calcified layer of cartilage and subchondral bone were better in the hACECM scaffold and hACECM scaffold + microfracture groups than the others, and excessive subchondral bone proliferation in the microfracture group. The results demonstrate that human cartilage extracellular matrix scaffold alone is an efficient, safe and simple way to repair cartilage defects. Conclusion hACECM scaffolds combined with/without microfracture facilitate chondral defect repair. The translational potential of this article Preclinical large animal models represent an important adjunct and surrogate for studies on articular cartilage repair, while the sheep stifle joint reflects many key features of the human knee and are therefore optimal experimental model for future clinical application in human. In this study, we developed a human articular cartilage-derived extracellular matrix scaffold and to verify the viability of its use in sheep animal models. Clinical studies are warranted to further quantify the effects of hACECM scaffolds in similar settings.
Collapse
Affiliation(s)
- Liqing Peng
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- Department of Orthopedics, First People's Hospital of Shuangliu District, No. 120, Chengbeishang Street, Shuangliu District, Chengdu, 610200, China
| | - Hao Li
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Haoyuan Deng
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Tianze Gao
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Runmeng Li
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Ziheng Xu
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Qinyu Tian
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Tianyuan Zhao
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Jianwei Li
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Yongkang Yang
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Chao Wang
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Shuyun Liu
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Quanyi Guo
- Institute of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin, 300071, China
| |
Collapse
|
17
|
Franco RAG, McKenna E, Shajib MS, Guillesser B, Robey PG, Crawford RW, Doran MR, Futrega K. Microtissue Culture Provides Clarity on the Relative Chondrogenic and Hypertrophic Response of Bone-Marrow-Derived Stromal Cells to TGF-β1, BMP-2, and GDF-5. Cells 2023; 13:37. [PMID: 38201241 PMCID: PMC10778331 DOI: 10.3390/cells13010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024] Open
Abstract
Chondrogenic induction of bone-marrow-derived stromal cells (BMSCs) is typically accomplished with medium supplemented with growth factors (GF) from the transforming growth factor-beta (TGF-β)/bone morphogenetic factor (BMP) superfamily. In a previous study, we demonstrated that brief (1-3 days) stimulation with TGF-β1 was sufficient to drive chondrogenesis and hypertrophy using small-diameter microtissues generated from 5000 BMSC each. This biology is obfuscated in typical large-diameter pellet cultures, which suffer radial heterogeneity. Here, we investigated if brief stimulation (2 days) of BMSC microtissues with BMP-2 (100 ng/mL) or growth/differentiation factor (GDF-5, 100 ng/mL) was also sufficient to induce chondrogenic differentiation, in a manner comparable to TGF-β1 (10 ng/mL). Like TGF-β1, BMP-2 and GDF-5 are reported to stimulate chondrogenic differentiation of BMSCs, but the effects of transient or brief use in culture have not been explored. Hypertrophy is an unwanted outcome in BMSC chondrogenic differentiation that renders engineered tissues unsuitable for use in clinical cartilage repair. Using three BMSC donors, we observed that all GFs facilitated chondrogenesis, although the efficiency and the necessary duration of stimulation differed. Microtissues treated with 2 days or 14 days of TGF-β1 were both superior at producing extracellular matrix and expression of chondrogenic gene markers compared to BMP-2 and GDF-5 with the same exposure times. Hypertrophic markers increased proportionally with chondrogenic differentiation, suggesting that these processes are intertwined for all three GFs. The rapid action, or "temporal potency", of these GFs to induce BMSC chondrogenesis was found to be as follows: TGF-β1 > BMP-2 > GDF-5. Whether briefly or continuously supplied in culture, TGF-β1 was the most potent GF for inducing chondrogenesis in BMSCs.
Collapse
Affiliation(s)
- Rose Ann G. Franco
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Eamonn McKenna
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Md. Shafiullah Shajib
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Bianca Guillesser
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Ross W. Crawford
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Michael R. Doran
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD 20892, USA
- Mater Research Institute—University of Queensland (UQ), Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Kathryn Futrega
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Schwarzl T, Keogh A, Shaw G, Krstic A, Clayton E, Higgins DG, Kolch W, Barry F. Transcriptional profiling of early differentiation of primary human mesenchymal stem cells into chondrocytes. Sci Data 2023; 10:758. [PMID: 37923731 PMCID: PMC10624874 DOI: 10.1038/s41597-023-02686-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 10/25/2023] [Indexed: 11/06/2023] Open
Abstract
Articular cartilage has only very limited regenerative capacities in humans. Tissue engineering techniques for cartilage damage repair are limited in the production of hyaline cartilage. Mesenchymal stem/stromal cells (MSCs) are multipotent stem cells and can be differentiated into mature cartilage cells, chondrocytes, which could be used for repairing damaged cartilage. Chondrogenesis is a highly complex, relatively inefficient process lasting over 3 weeks in vitro. Methods: In order to better understand chondrogenic differentiation, especially the commitment phase, we have performed transcriptional profiling of MSC differentiation into chondrocytes from early timepoints starting 15 minutes after induction to 16 hours and fully differentiated chondrocytes at 21 days in triplicates.
Collapse
Affiliation(s)
- Thomas Schwarzl
- European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Andrea Keogh
- Previously: Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland
| | - Georgina Shaw
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland
| | - Aleksandar Krstic
- Systems Biology Ireland (SBI), School of Medicine, University College Dublin, Dublin, 4, Ireland
| | - Elizabeth Clayton
- Previously: Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland
| | - Desmond G Higgins
- Systems Biology Ireland (SBI), School of Medicine, University College Dublin, Dublin, 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Belfield, Dublin, 4, Ireland
| | - Walter Kolch
- Systems Biology Ireland (SBI), School of Medicine, University College Dublin, Dublin, 4, Ireland.
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Belfield, Dublin, 4, Ireland.
| | - Frank Barry
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland
| |
Collapse
|
19
|
Fani N, Peshkova M, Bikmulina P, Golroo R, Timashev P, Vosough M. Fabricating the cartilage: recent achievements. Cytotechnology 2023; 75:269-292. [PMID: 37389132 PMCID: PMC10299965 DOI: 10.1007/s10616-023-00582-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 05/09/2023] [Indexed: 07/01/2023] Open
Abstract
This review aims to describe the most recent achievements and provide an insight into cartilage engineering and strategies to restore the cartilage defects. Here, we discuss cell types, biomaterials, and biochemical factors applied to form cartilage tissue equivalents and update the status of fabrication techniques, which are used at all stages of engineering the cartilage. The actualized concept to improve the cartilage tissue restoration is based on applying personalized products fabricated using a full cycle platform: a bioprinter, a bioink consisted of ECM-embedded autologous cell aggregates, and a bioreactor. Moreover, in situ platforms can help to skip some steps and enable adjusting the newly formed tissue in the place during the operation. Only some achievements described have passed first stages of clinical translation; nevertheless, the number of their preclinical and clinical trials is expected to grow in the nearest future.
Collapse
Affiliation(s)
- Nesa Fani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Polina Bikmulina
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, Moscow, Russia
| | - Reihaneh Golroo
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
20
|
Kim KI, Lee MC, Lee JH, Moon YW, Lee WS, Lee HJ, Hwang SC, In Y, Shon OJ, Bae KC, Song SJ, Park KK, Kim JH. Clinical Efficacy and Safety of the Intra-articular Injection of Autologous Adipose-Derived Mesenchymal Stem Cells for Knee Osteoarthritis: A Phase III, Randomized, Double-Blind, Placebo-Controlled Trial. Am J Sports Med 2023; 51:2243-2253. [PMID: 37345256 DOI: 10.1177/03635465231179223] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/23/2023]
Abstract
BACKGROUND Intra-articular injection of autologous culture-expanded adipose-derived mesenchymal stem cells (ADMSCs) has introduced a promising treatment option for knee osteoarthritis. Although the clinical efficacy and safety of ADMSCs have been reported, the treatment remains controversial owing to the small sample sizes and heterogeneous osteoarthritis grades in previous studies. PURPOSE To assess the efficacy and safety of intra-articular injection of ADMSCs as compared with placebo in alleviating pain and improving functional capacity in a large sample of patients with knee osteoarthritis of Kellgren-Lawrence (K-L) grade 3. STUDY DESIGN Randomized controlled trial; Level of evidence, 1. METHODS This phase III multicenter clinical trial was a double-blind randomized controlled study that included 261 patients with K-L grade 3 symptomatic knee osteoarthritis who were administered a single injection of autologous culture-expanded ADMSCs or placebo. Clinical data were assessed at baseline and at 3 and 6 months after the injection. The primary endpoints were improvements in 100-mm visual analog scale (VAS) for pain and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) for function at 6 months after the injection. The secondary endpoints included clinical and radiologic examinations and safety after injection. The changes in cartilage defects after injection were assessed by magnetic resonance imaging at 6 months. RESULTS The ADMSC and control groups included 125 and 127 patients available for follow-up, respectively. At 6 months, the ADMSC group showed significantly better improvements in 100-mm VAS (ADMSC vs control, 25.2 vs 15.5; P = .004) and total WOMAC score (21.7 vs 14.3; P = .002) as compared with the control group. The linear mixed model analysis indicated significantly better improvements in all clinical outcomes in the ADMSC group after 6 months. At 6 months, the ADMSC group achieved significantly higher proportions of patients above the minimal clinically important difference in 100-mm VAS and WOMAC score. Radiologic outcomes and adverse events did not demonstrate significant differences between the groups. No serious treatment-related adverse events were observed. Magnetic resonance imaging revealed no significant difference in change of cartilage defects between the groups at 6 months. CONCLUSION Intra-articular injection of autologous culture-expanded ADMSCs provided significant pain relief and functional improvements in patients with K-L grade 3 osteoarthritis. Long-term results are needed to determine the disease-modifying effects of ADMSCs, such as structural changes, and the duration of effect of intra-articular injection of ADMSCs in knee osteoarthritis. REGISTRATION NCT03990805 (ClinicalTrials.gov identifier).
Collapse
Affiliation(s)
- Kang-Il Kim
- Department of Orthopaedic Surgery, Center for Joint Diseases, Kyung Hee University Hospital at Gangdong, Seoul, Korea; Department of Orthopaedic Surgery, School of Medicine, Kyung Hee University, Seoul, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Myung Chul Lee
- Department of Orthopedic Surgery, Seoul National University Hospital, Seoul National University School of Medicine, Seoul, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Ju Hong Lee
- Department of Orthopaedic Surgery, Jeonbuk National University Hospital, Jeonbuk National University School of Medicine, Jeonju, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Young-Wan Moon
- Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Woo-Suk Lee
- Department of Orthopedic Surgery, Gangnam Severance Hospital, Yonsei University School of Medicine, Seoul, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Han-Jun Lee
- Department of Orthopedic Surgery, Chung-Ang University Hospital, Chung-Ang University School of Medicine, Seoul, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Sun-Chul Hwang
- Department of Orthopaedic Surgery, Institute of Health Science, Research Institute of Life Science, and School of Medicine, Gyeongsang National University, Jinju, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Yong In
- Department of Orthopaedic Surgery, Seoul St Mary's Hospital, Catholic University College of Medicine, Seoul, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Oog-Jin Shon
- Department of Orthopaedic Surgery, Yeungnam University Medical Center, Yeungnam University School of Medicine, Daegu, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Ki-Cheor Bae
- Department of Orthopaedic Surgery, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Sang-Jun Song
- Department of Orthopaedic Surgery, Kyung Hee University Medical Center, Kyung Hee University School of Medicine, Seoul, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Kwan Kyu Park
- Department of Orthopedic Surgery, Severance Hospital, Yonsei University School of Medicine, Seoul, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Jun-Ho Kim
- Department of Orthopaedic Surgery, Center for Joint Diseases, Kyung Hee University Hospital at Gangdong, Seoul, Korea
- Investigation performed at Kyung Hee University Hospital at Gangdong, Seoul, Korea
| |
Collapse
|
21
|
Arhebamen EP, Teodoro MT, Blonka AB, Matthew HWT. Long-Term Culture Performance of a Polyelectrolyte Complex Microcapsule Platform for Hyaline Cartilage Repair. Bioengineering (Basel) 2023; 10:bioengineering10040467. [PMID: 37106654 PMCID: PMC10135885 DOI: 10.3390/bioengineering10040467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Articular cartilage (AC) tissue repair and regeneration remains an ongoing challenge. One component of the challenge is the limited ability to scale an engineered cartilage graft to clinically relevant sizes while maintaining uniform properties. In this paper, we report on the evaluation of our polyelectrolyte complex microcapsule (PECM) platform technology as a technique for generating cartilage-like spherical modules. Bone marrow-derived mesenchymal stem cells (bMSCs) or primary articular chondrocytes were encapsulated within PECMs composed of methacrylated hyaluronan, collagen I, and chitosan. The formation of cartilage-like tissue in the PECMs over a 90-day culture was characterized. The results showed that chondrocytes exhibited superior growth and matrix deposition compared to either chondrogenically-induced bMSCs or a mixed PECM culture containing both chondrocytes and bMSCs. The chondrocyte-generated matrix filled the PECM and produced substantial increases in capsule compressive strength. The PECM system thus appears to support intracapsular cartilage tissue formation and the capsule approach promotes efficient culture and handling of these micro tissues. Since previous studies have proven the feasibility of fusing such capsules into large tissue constructs, the results suggest that encapsulating primary chondrocytes in PECM modules may be a viable route toward achieving a functional articular cartilage graft.
Collapse
Affiliation(s)
- Ehinor P Arhebamen
- Department of Biomedical Engineering, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
| | - Maria T Teodoro
- Department of Biomedical Engineering, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
| | - Amelia B Blonka
- Department of Biomedical Engineering, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
| | - Howard W T Matthew
- Department of Biomedical Engineering, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
- Department of Chemical Engineering and Materials Science, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
| |
Collapse
|
22
|
Goh D, Yang Y, Lee EH, Hui JHP, Yang Z. Managing the Heterogeneity of Mesenchymal Stem Cells for Cartilage Regenerative Therapy: A Review. Bioengineering (Basel) 2023; 10:bioengineering10030355. [PMID: 36978745 PMCID: PMC10045936 DOI: 10.3390/bioengineering10030355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/12/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Articular cartilage defects commonly result from trauma and are associated with significant morbidity. Since cartilage is an avascular, aneural, and alymphatic tissue with a poor intrinsic healing ability, the regeneration of functional hyaline cartilage remains a difficult clinical problem. Mesenchymal stem cells (MSCs) are multipotent cells with multilineage differentiation potential, including the ability to differentiate into chondrocytes. Due to their availability and ease of ex vivo expansion, clinicians are increasingly applying MSCs in the treatment of cartilage lesions. However, despite encouraging pre-clinical and clinical data, inconsistencies in MSC proliferative and chondrogenic potential depending on donor, tissue source, cell subset, culture conditions, and handling techniques remain a key barrier to widespread clinical application of MSC therapy in cartilage regeneration. In this review, we highlight the strategies to manage the heterogeneity of MSCs ex vivo for more effective cartilage repair, including reducing the MSC culture expansion period, and selecting MSCs with higher chondrogenic potential through specific genetic markers, surface markers, and biophysical attributes. The accomplishment of a less heterogeneous population of culture-expanded MSCs may improve the scalability, reproducibility, and standardisation of MSC therapy for clinical application in cartilage regeneration.
Collapse
Affiliation(s)
- Doreen Goh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Yanmeng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
- Correspondence: ; Tel.: +65-6516-5398
| |
Collapse
|
23
|
Tolabi H, Davari N, Khajehmohammadi M, Malektaj H, Nazemi K, Vahedi S, Ghalandari B, Reis RL, Ghorbani F, Oliveira JM. Progress of Microfluidic Hydrogel-Based Scaffolds and Organ-on-Chips for the Cartilage Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2208852. [PMID: 36633376 DOI: 10.1002/adma.202208852] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/09/2022] [Indexed: 05/09/2023]
Abstract
Cartilage degeneration is among the fundamental reasons behind disability and pain across the globe. Numerous approaches have been employed to treat cartilage diseases. Nevertheless, none have shown acceptable outcomes in the long run. In this regard, the convergence of tissue engineering and microfabrication principles can allow developing more advanced microfluidic technologies, thus offering attractive alternatives to current treatments and traditional constructs used in tissue engineering applications. Herein, the current developments involving microfluidic hydrogel-based scaffolds, promising structures for cartilage regeneration, ranging from hydrogels with microfluidic channels to hydrogels prepared by the microfluidic devices, that enable therapeutic delivery of cells, drugs, and growth factors, as well as cartilage-related organ-on-chips are reviewed. Thereafter, cartilage anatomy and types of damages, and present treatment options are briefly overviewed. Various hydrogels are introduced, and the advantages of microfluidic hydrogel-based scaffolds over traditional hydrogels are thoroughly discussed. Furthermore, available technologies for fabricating microfluidic hydrogel-based scaffolds and microfluidic chips are presented. The preclinical and clinical applications of microfluidic hydrogel-based scaffolds in cartilage regeneration and the development of cartilage-related microfluidic chips over time are further explained. The current developments, recent key challenges, and attractive prospects that should be considered so as to develop microfluidic systems in cartilage repair are highlighted.
Collapse
Affiliation(s)
- Hamidreza Tolabi
- New Technologies Research Center (NTRC), Amirkabir University of Technology, Tehran, 15875-4413, Iran
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, 15875-4413, Iran
| | - Niyousha Davari
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran
| | - Mehran Khajehmohammadi
- Department of Mechanical Engineering, Faculty of Engineering, Yazd University, Yazd, 89195-741, Iran
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, 8916877391, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg, 9220, Denmark
| | - Katayoun Nazemi
- Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Samaneh Vahedi
- Department of Material Science and Engineering, Faculty of Engineering, Imam Khomeini International University, Qazvin, 34149-16818, Iran
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4805-017, Portugal
| | - Farnaz Ghorbani
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058, Erlangen, Germany
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4805-017, Portugal
| |
Collapse
|
24
|
Franco RAG, McKenna E, Robey PG, Crawford RW, Doran MR, Futrega K. SP7 gene silencing dampens bone marrow stromal cell hypertrophy, but it also dampens chondrogenesis. J Tissue Eng 2023; 14:20417314231177136. [PMID: 37362901 PMCID: PMC10288420 DOI: 10.1177/20417314231177136] [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: 02/11/2023] [Accepted: 05/05/2023] [Indexed: 06/28/2023] Open
Abstract
For bone marrow stromal cells (BMSC) to be useful in cartilage repair their propensity for hypertrophic differentiation must be overcome. A single day of TGF-β1 stimulation activates intrinsic signaling cascades in BMSCs which subsequently drives both chondrogenic and hypertrophic differentiation. TGF-β1 stimulation upregulates SP7, a transcription factor known to contribute to hypertrophic differentiation, and SP7 remains upregulated even if TGF-β1 is subsequently withdrawn from the chondrogenic induction medium. Herein, we stably transduced BMSCs to express an shRNA designed to silence SP7, and assess the capacity of SP7 silencing to mitigate hypertrophy. SP7 silencing dampened both hypertrophic and chondrogenic differentiation processes, resulting in diminished microtissue size, impaired glycosaminoglycan production and reduced chondrogenic and hypertrophic gene expression. Thus, while hypertrophic features were dampened by SP7 silencing, chondrogenic differentation was also compromised. We further investigated the role of SP7 in monolayer osteogenic and adipogenic cultures, finding that SP7 silencing dampened characteristic mineralization and lipid vacuole formation, respectively. Overall, SP7 silencing affects the trilineage differentiation of BMSCs, but is insufficient to decouple BMSC hypertrophy from chondrogenesis. These data highlight the challenge of promoting BMSC chondrogenesis whilst simultaneously reducing hypertrophy in cartilage tissue engineering strategies.
Collapse
Affiliation(s)
- Rose Ann G Franco
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Translational Research Institute (TRI), Brisbane, QLD, Australia
- Center for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Eamonn McKenna
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Translational Research Institute (TRI), Brisbane, QLD, Australia
- Center for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Pamela G Robey
- Skeletal Biology Section (SBS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| | - Ross W Crawford
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Center for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Michael R Doran
- Translational Research Institute (TRI), Brisbane, QLD, Australia
- Center for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Skeletal Biology Section (SBS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- AstraZeneca, Biologics Engineering, Oncology R&D, One MedImmune Way, Gaithersburg, MD, USA
| | - Kathryn Futrega
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Translational Research Institute (TRI), Brisbane, QLD, Australia
- Center for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Skeletal Biology Section (SBS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| |
Collapse
|
25
|
Kutaish H, Tscholl PM, Cosset E, Bengtsson L, Braunersreuther V, Mor FM, Laedermann J, Furfaro I, Stafylakis D, Hannouche D, Gerstel E, Krause KH, Assal M, Menetrey J, Tieng V. Articular Cartilage Repair After Implantation of Hyaline Cartilage Beads Engineered From Adult Dedifferentiated Chondrocytes: Cartibeads Preclinical Efficacy Study in a Large Animal Model. Am J Sports Med 2023; 51:237-249. [PMID: 36592016 DOI: 10.1177/03635465221138099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Chondrocyte-based cell therapy to repair cartilage has been used for >25 years despite current limitations. This work presents a new treatment option for cartilage lesions. HYPOTHESIS High-quality hyaline cartilage microtissues called Cartibeads are capable of treating focal chondral lesions once implanted in the defect, by complete fusion of Cartibeads among themselves and their integration with the surrounding native cartilage and subchondral bone. STUDY DESIGN Controlled laboratory study. METHODS Cartibeads were first produced from human donors and characterized using histology (safranin O staining of glycosaminoglycan [GAG] and immunohistochemistry of collagen I and II) and GAG dosage. Cartibeads from 6 Göttingen minipigs were engineered and implanted in an autologous condition in the knee (4 or 5 lesions per knee). One group was followed up for 3 months and the other for 6 months. Feasibility and efficacy were measured using histological analysis and macroscopic and microscopic scores. RESULTS Cartibeads revealed hyaline features with strong staining of GAG and collagen II. High GAG content was obtained: 24.6-µg/mg tissue (wet weight), 15.52-µg/mg tissue (dry weight), and 35 ± 3-µg GAG/bead (mean ± SD). Histological analysis of Göttingen minipigs showed good integration of Cartibeads grafts at 3 and 6 months after implantation. The Bern Score of the histological assay comparing grafted versus empty lesions was significant at 3 months (grafted, n = 10; nongrafted, n = 4; score, 3.3 and 5.3, respectively) and 6 months (grafted, n = 11; nongrafted, n = 3; score, 1.6 and 5.1). CONCLUSION We developed an innovative 3-step method allowing, for the first time, the use of fully dedifferentiated adult chondrocytes with a high number of cell passage (owing to the extensive amplification in culture). Cartibeads engineered from chondrocytes hold potential as an advanced therapy medicinal product for treating cartilage lesions with established efficacy. CLINICAL RELEVANCE This successful preclinical study, combined with standardized manufacturing of Cartibeads according to good manufacturing practice guidelines, led to the approval of first-in-human clinical trial by the ethics committee and local medical authority. The generated data highlighted a promising therapy to treat cartilage lesions from a small amount of starting biopsy specimen. With our innovative cell amplification technology, very large lesions can be treated, and older active patients can benefit from it.
Collapse
Affiliation(s)
- Halah Kutaish
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland. University Medical Center, University of Geneva, Geneva, Switzerland. Foot and Ankle Surgery Centre, Centre Assal, Clinique La Colline, Hirslanden Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Matthias Tscholl
- Department of Orthopaedics Surgery, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Erika Cosset
- University Medical Center, University of Geneva, Geneva, Switzerland. Laboratory of Tumor Immunology, Oncology Department, Center for Translational Research in Onco- Hematology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Laura Bengtsson
- University Medical Center, University of Geneva, Geneva, Switzerland. Vanarix SA, Lausanne, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Vincent Braunersreuther
- Service of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Flavio Maurizio Mor
- Tissue Engineering Laboratory, HEPIA/HES-SO, University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Jeremy Laedermann
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Ivan Furfaro
- Laboratory for Soft Bioelectronic Interfaces, Institute of Microengineering, Institute of Bioengineering, Centre for Neuroprosthetics, École Polytechnique Fédeérale de Lausanne (EPFL), Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Dimitrios Stafylakis
- Department of Orthopaedics Surgery, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Didier Hannouche
- University Medical Center, University of Geneva, Geneva, Switzerland. Department of Orthopaedics Surgery, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Eric Gerstel
- University Medical Center, University of Geneva, Geneva, Switzerland. Clinique la Colline, Hirslanden, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland. University Medical Center, University of Geneva, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Mathieu Assal
- University Medical Center, University of Geneva, Geneva, Switzerland. Foot and Ankle Surgery Centre, Centre Assal, Clinique La Colline, Hirslanden Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Jacques Menetrey
- University Medical Center, University of Geneva, Geneva, Switzerland. Centre for Sports Medicine and Exercise, Clinique la Colline, Hirslanden, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Vannary Tieng
- University Medical Center, University of Geneva, Geneva, Switzerland. Vanarix SA, Lausanne, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
26
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
27
|
Kutaish H, Bengtsson L, Tscholl PM, Marteyn A, Braunersreuther V, Guérin A, Béna F, Gimelli S, Longet D, Ilmjärv S, Dietrich PY, Gerstel E, Jaquet V, Hannouche D, Menetrey J, Assal M, Krause KH, Cosset E, Tieng V. Hyaline Cartilage Microtissues Engineered from Adult Dedifferentiated Chondrocytes: Safety and Role of WNT Signaling. Stem Cells Transl Med 2022; 11:1219-1231. [PMID: 36318262 PMCID: PMC9801297 DOI: 10.1093/stcltm/szac074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/18/2022] [Indexed: 11/05/2022] Open
Abstract
The repair of damaged articular cartilage is an unmet medical need. Chondrocyte-based cell therapy has been used to repair cartilage for over 20 years despite current limitations. Chondrocyte dedifferentiation upon expansion in monolayer is well known and is the main obstacle to their use as cell source for cartilage repair. Consequently, current approaches often lead to fibrocartilage, which is biomechanically different from hyaline cartilage and not effective as a long-lasting treatment. Here, we describe an innovative 3-step method to engineer hyaline-like cartilage microtissues, named Cartibeads, from high passage dedifferentiated chondrocytes. We show that WNT5A/5B/7B genes were highly expressed in dedifferentiated chondrocytes and that a decrease of the WNT signaling pathway was instrumental for full re-differentiation of chondrocytes, enabling production of hyaline matrix instead of fibrocartilage matrix. Cartibeads showed hyaline-like characteristics based on GAG quantity and type II collagen expression independently of donor age and cartilage quality. In vivo, Cartibeads were not tumorigenic when transplanted into SCID mice. This simple 3-step method allowed a standardized production of hyaline-like cartilage microtissues from a small cartilage sample, making Cartibeads a promising candidate for the treatment of cartilage lesions.
Collapse
Affiliation(s)
| | | | - Philippe Matthias Tscholl
- University Medical Center, University of Geneva, Geneva, Switzerland,Department of Orthopaedics Surgery, Geneva University Hospital, Geneva, Switzerland
| | - Antoine Marteyn
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | - Vincent Braunersreuther
- Service of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Alexandre Guérin
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | - Frédérique Béna
- Service of Genetic Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Stefania Gimelli
- Service of Genetic Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - David Longet
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | - Sten Ilmjärv
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | - Pierre-Yves Dietrich
- Laboratory of Tumor Immunology, Oncology Department, Center for Translational Research in Onco-Hematology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Eric Gerstel
- University Medical Center, University of Geneva, Geneva, Switzerland,Clinique la Colline, Hirslanden, Geneva, Switzerland
| | - Vincent Jaquet
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland,READS Unit, Medical School, University of Geneva, Geneva, Switzerland
| | - Didier Hannouche
- University Medical Center, University of Geneva, Geneva, Switzerland,Department of Orthopaedics Surgery, Geneva University Hospital, Geneva, Switzerland
| | - Jacques Menetrey
- University Medical Center, University of Geneva, Geneva, Switzerland,Centre for Sports Medicine and Exercise, Clinique la Colline, Hirslanden, Geneva, Switzerland
| | - Mathieu Assal
- University Medical Center, University of Geneva, Geneva, Switzerland,Foot and Ankle Surgery Centre, Centre Assal, Clinique La Colline, Hirslanden Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland,University Medical Center, University of Geneva, Geneva, Switzerland
| | | | - Vannary Tieng
- Corresponding author: Vannary Tieng, Vanarix SA, Avenue Mon-Repos 14, 1005 Lausanne, Switzerland.
| |
Collapse
|
28
|
Decoene I, Herpelinck T, Geris L, Luyten FP, Papantoniou I. Engineering bone-forming callus organoid implants in a xenogeneic-free differentiation medium. FRONTIERS IN CHEMICAL ENGINEERING 2022. [DOI: 10.3389/fceng.2022.892190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The field of tissue engineering aspires to provide clinically relevant solutions for patients through the integration of developmental engineering principles with a bottom-up manufacturing approach. However, the manufacturing of cell-based advanced therapy medicinal products is hampered by protocol complexity, lack of non-invasive critical quality controls, and dependency on animal-derived components for tissue differentiation. We investigate a serum-free, chemically defined, xeno- and lipid-free chondrogenic differentiation medium to generate bone-forming callus organoids. Our results show an increase in microtissue homogeneity during prolonged differentiation and the high quality of in vivo bone-forming organoids. The low protein content of the culture medium potentially allows for the monitoring of relevant secreted biomarkers as (critical) quality attributes. Together, we envisage that this xeno- and lipid-free chondrogenic medium is compatible with industrial scale-up and automation while facilitating the implementation of non-invasive imaging and the use of quality control parameters based on secreted biomarkers.
Collapse
|
29
|
Kim JH, Kim KI, Yoon WK, Song SJ, Jin W. Intra-articular Injection of Mesenchymal Stem Cells After High Tibial Osteotomy in Osteoarthritic Knee: Two-Year Follow-up of Randomized Control Trial. Stem Cells Transl Med 2022; 11:572-585. [PMID: 35674255 PMCID: PMC9216209 DOI: 10.1093/stcltm/szac023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 03/20/2022] [Indexed: 02/02/2023] Open
Abstract
Intra-articular injection of adipose-derived mesenchymal stem cell (ADMSC) after medial open-wedge high tibial osteotomy (MOWHTO) would be a promising disease-modifying treatment by correcting biomechanical and biochemical environment for arthritic knee with varus malalignment. However, there is a paucity of clinical evidence of the treatment. This randomized controlled trial (RCT) was aimed to assess regeneration of cartilage defect, functional improvement, and safety of intra-articular injection of ADMSCs after MOWHTO compared with MOWHTO alone for osteoarthritic knee with varus malalignment. This RCT allocated 26 patients into the MOWHTO with ADMSC-injection group (n = 13) and control (MOWHTO-alone) group (n = 13). The primary outcome was the serial changes of cartilage defect on periodic magnetic resonance imaging (MRI) evaluation using valid measurements until postoperative 24 months. Secondary outcomes were the 2-stage arthroscopic evaluation for macroscopic cartilage status and the postoperative functional improvements of patient-reported outcome measures until the latest follow-up. Furthermore, safety profiles after the treatment were evaluated. Cartilage regeneration on serial MRIs showed significantly better in the ADMSC group than in the control group. The arthroscopic assessment revealed that total cartilage regeneration was significantly better in the ADMSC group. Although it was not significant, functional improvements after the treatment showed a tendency to be greater in the ADMSC group than in the control group from 18 months after the treatment. No treatment-related adverse events, serious adverse events, and postoperative complications occurred in all cases. Concomitant intra-articular injection of ADMSCs with MOWHTO had advantages over MOWHTO alone in terms of cartilage regeneration with safety at 2-year follow-up, suggesting potential disease-modifying treatment for knee OA with varus malalignment.
Collapse
Affiliation(s)
- Jun-Ho Kim
- Department of Orthopaedic Surgery, Center for Joint Diseases, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Kang-Il Kim
- Corresponding author: Kang-Il Kim, M.D., Ph D., Department of Orthopedic Surgery, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-gu, Seoul 134-727, Republic of Korea. Tel: +82 2 440 6151;
| | - Wan Keun Yoon
- Department of Orthopaedic Surgery, Center for Joint Diseases, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Sang-Jun Song
- Department of Orthopaedic Surgery, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Wook Jin
- Department of Radiology, Kyung Hee University Hospital at Gandong, Seoul, Republic of Korea
| |
Collapse
|
30
|
Kim J, Park J, Song SY, Kim E. Advanced Therapy medicinal products for autologous chondrocytes and comparison of regulatory systems in target countries. Regen Ther 2022; 20:126-137. [PMID: 35582708 PMCID: PMC9079100 DOI: 10.1016/j.reth.2022.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/27/2022] [Accepted: 04/14/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction Autologous chondrocytes (ACs) are Human cell/tissue-based products used for the treatment of joint cartilage defects. Regulatory agencies have established regulations related to ACs to ensure their safety and efficacy. This study investigated the status and characteristics of ACs approved worldwide. Furthermore, the AC-related regulations were compared by country to provide reference materials for the development of product approval procedures. Methods This study reviewed the current status of global AC products over the past 20 years by referring to the AC approval list provided on the International Society for Cell & Gene Therapy (ISCT) website. Based on the review report provided by the regulatory agencies that approved the products, major nonclinical/clinical data and product characteristics were reviewed; and the classification and definition of ACs and the approval review procedures were compared through the regulatory agencies’ websites. The development status of ACs was also analyzed using a clinical trial registration site. Results Eight ACs were approved during the study period in Europe, the US, Japan, Australia, and Korea. Two products were withdrawn owing to marketability problems. Human cell/tissue-based products in each country are classified and defined distinguished from biopharmaceuticals, but the approval process for both products is the same. The approval period differs by country, with an average of 282.4 days and the shortest being in Korea (115 days). On Clinical Trials.gov, we screened 46 clinical trials related to ACs, which were conducted in Europe (41%), Korea (20%), and the US (17%). The knee accounted for the largest portion of the indication (37/46, 80%), followed by the ankle or hip joints. Measurements of improvements in function and pain were the main endpoints used to evaluate the efficacy of ACs. Observational studies were conducted to confirm the long-term safety of these products. Conclusions This is the first study comparing the current status and characteristics of globally approved AC products, as well as their classification and definition by country. In the past two decades, clinical trials have been conducted on the application of ACs in tissue engineering to treat joint cartilage defects. ACs are expected to be used for the treatment of cartilage defect diseases. This is the first study that compared AC products that are currently approved globally per country. AC products are classified distinguished from biopharmaceuticals. Regulatory agencies implement systems to ensure long-term safety and efficacy of ACs.
Collapse
Affiliation(s)
- Jiwon Kim
- Department of Pharmaceutical Industry, Chung-Ang University, Seoul, 06974, Republic of Korea
- Data Science, Evidence-Based Clinical Research Laboratory, Departments of Health Science & Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jaehong Park
- Department of Pharmaceutical Industry, Chung-Ang University, Seoul, 06974, Republic of Korea
- Data Science, Evidence-Based Clinical Research Laboratory, Departments of Health Science & Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seung-Yeon Song
- Data Science, Evidence-Based Clinical Research Laboratory, Departments of Health Science & Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Eunyoung Kim
- Department of Pharmaceutical Industry, Chung-Ang University, Seoul, 06974, Republic of Korea
- Data Science, Evidence-Based Clinical Research Laboratory, Departments of Health Science & Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
- Corresponding author. Division of Licensing of Medicines and Regulatory Science, The Graduate School of Pharmaceutical Management, Chung-Ang University, 84 Heukseok-Ro, Dangjak-gu, Seoul, 06974, Republic of Korea. Tel: +82-2-820-5791, Fax: +82-2-816-7338.
| |
Collapse
|
31
|
Franco RAG, McKenna E, Robey PG, Shajib MS, Crawford RW, Doran MR, Futrega K. Inhibition of BMP signaling with LDN 193189 can influence bone marrow stromal cell fate but does not prevent hypertrophy during chondrogenesis. Stem Cell Reports 2022; 17:616-632. [PMID: 35180395 PMCID: PMC9039850 DOI: 10.1016/j.stemcr.2022.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/22/2023] Open
Abstract
Bone morphogenetic protein (BMP) cascades are upregulated during bone marrow-derived stromal cell (BMSC) chondrogenesis, contributing to hypertrophy and preventing effective BMSC-mediated cartilage repair. Previous work demonstrated that a proprietary BMP inhibitor prevented BMSC hypertrophy, yielding stable cartilage tissue. Because of the significant therapeutic potential of a molecule capable of hypertrophy blockade, we evaluated the capacity of a commercially available BMP type I receptor inhibitor with similar properties, LDN 193189, to prevent BMSC hypertrophy. Using 14-day microtissue chondrogenic induction cultures we found that LDN 193189 permitted BMSC chondrogenesis but did not prevent hypertrophy. LDN 193189 was sufficiently potent to counter mineralization and adipogenesis in response to exogenous BMP-2 in osteogenic induction cultures. LDN 193189 did not modify BMSC behavior in adipogenic induction cultures. Although LDN 193189 is effective in countering BMP signaling in a manner that influences BMSC fate, this blockade is insufficient to prevent hypertrophy.
Collapse
Affiliation(s)
- Rose Ann G Franco
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia
| | - Eamonn McKenna
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia
| | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Md Shaffiulah Shajib
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Ross W Crawford
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia
| | - Michael R Doran
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia; Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Mater Research Institute - University of Queensland, Brisbane, Australia.
| | - Kathryn Futrega
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia; Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA.
| |
Collapse
|
32
|
Wang S, Yang L, Cai B, Liu F, Hou Y, Zheng H, Cheng F, Zhang H, Wang L, Wang X, Lv Q, Kong L, Lee KB, Zhang Q. Injectable hybrid inorganic nanoscaffold-templated rapid stem cell assembly for cartilage repair. Natl Sci Rev 2022; 9:nwac037. [PMID: 35419207 PMCID: PMC8998491 DOI: 10.1093/nsr/nwac037] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 11/14/2022] Open
Abstract
ABSTRACT
Cartilage injuries are often devastating and most cannot be cured because of the intrinsically low regenerative capacity of cartilage tissues. Although stem cell therapy has shown enormous potential for cartilage repair, the therapeutic outcome has been restricted by low survival rates and poor chondrocyte differentiation in vivo. Here, we report an injectable hybrid inorganic (IHI) nanoscaffold that facilitates fast assembly, enhances survival, and regulates chondrogenic differentiation of stem cells. IHI nanoscaffolds that strongly bind to extracellular matrix (ECM) proteins assemble stem cells through synergistic three-dimensional (3D) cell-cell and cell-matrix interactions, creating a favourable physical microenvironment for stem cell survival and differentiation in vitro and in vivo. Additionally, chondrogenic factors can be loaded into nanoscaffolds with a high capacity, which allows deep, homogenous drug delivery into assembled 3D stem cell-derived tissues for effective control over the soluble microenvironment of stem cells. The developed IHI nanoscaffolds that assemble with stem cells are injectable. They also scavenge reactive oxygen species and timely biodegrade for proper integration into injured cartilage tissues. Implantation of stem cell-assembled IHI nanoscaffolds into injured cartilage results in accelerated tissue regeneration and functional recovery. By establishing our IHI nanoscaffold-templated 3D stem cell assembly method, we provide a promising approach to better overcome the inhibitory microenvironment associated with cartilage injuries and to advance current stem cell-based tissue engineering.
Collapse
Affiliation(s)
- Shenqiang Wang
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
- Xi’an Key Laboratory of Functional Organic Porous Materials, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bolei Cai
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Fuwei Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Hua Zheng
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
| | - Fang Cheng
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
| | - Hepeng Zhang
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
- Xi’an Key Laboratory of Functional Organic Porous Materials, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| | - Le Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Xiaoyi Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Qianxin Lv
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Liang Kong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Qiuyu Zhang
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
- Xi’an Key Laboratory of Functional Organic Porous Materials, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| |
Collapse
|
33
|
Sun K, Tao C, Wang DA. Scaffold-free approaches for the fabrication of engineered articular cartilage tissue. Biomed Mater 2022; 17. [PMID: 35114657 DOI: 10.1088/1748-605x/ac51b9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/03/2022] [Indexed: 11/12/2022]
Abstract
Tissue engineered cartilaginous constructs have meet great advances in the past decades as a treatment for osteoarthritis, a degenerative disease affecting people all over the world as the population ages. Scaffold-free tissue engineered constructs are designed and developed in recent years with only cells and cell-derived matrix involved. Scaffold-free tissue constructs do not require cell adherence on exogenous materials and are superior to scaffold-based constructs in (1) relying on only cells to produce matrix, (2) not interfering cell-cell signaling, cell migration or small molecules diffusion after implantation and (3) introducing no exogenous impurities. In this review, three main scaffold-free methodologies for cartilage tissue engineering, the cell sheet technology, the phase transfer cell culture-living hyaline cartilage graft (PTCC-LhCG) system and the cell aggregate-based (bottom-up) methods, were reviewed, covering mold fabrication, decellularization and 3D bioprinting. The recent advances, medical applications, superiority and drawbacks were elaborated in detail.
Collapse
Affiliation(s)
- Kang Sun
- City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Kowloon, 000000, HONG KONG
| | - Chao Tao
- City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Kowloon, 000000, HONG KONG
| | - Dong-An Wang
- City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Kowloon, 000000, HONG KONG
| |
Collapse
|
34
|
Li M, Sun D, Zhang J, Wang Y, Wei Q, Wang Y. Application and development of 3D bioprinting in cartilage tissue engineering. Biomater Sci 2022; 10:5430-5458. [DOI: 10.1039/d2bm00709f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bioprinting technology can build complex tissue structures and has the potential to fabricate engineered cartilage with bionic structures for achieving cartilage defect repair/regeneration.
Collapse
Affiliation(s)
- Mingyang Li
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Daocen Sun
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Juan Zhang
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yanmei Wang
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Qinghua Wei
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yanen Wang
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
35
|
Ray S, Chavan C. Current scenario of clinical trials on stem cells as a drug in India: A clinical trials registry of India database analysis. Perspect Clin Res 2022. [DOI: 10.4103/picr.picr_140_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
36
|
Kim KI, Lee WS, Kim JH, Bae JK, Jin W. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:586-596. [PMID: 35567774 PMCID: PMC9216498 DOI: 10.1093/stcltm/szac024] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/20/2022] [Indexed: 11/15/2022] Open
Abstract
Although successful short-term results of the intra-articular injection of mesenchymal stem cells (MSCs) for the conservative treatment of knee osteoarthritis (OA) have been reported, the mid-term results of the injection of adipose-derived (AD) MSCs remains unknown. We assessed the mid-term safety and efficacy of the intra-articular injection of ADMSCs in patients with knee OA. Eleven patients with knee OA were prospectively enrolled and underwent serial evaluations during a 5-year follow-up of a single intra-articular injection of autologous high-dose (1.0 × 108) ADMSCs. The safety profiles were assessed using the World Health Organization Common Toxicity Criteria. The clinical evaluations included visual analog scale (VAS) and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores for pain and function, respectively. The radiologic evaluations included chondral defect area and whole-organ magnetic resonance imaging scores (WORMS) by serial magnetic resonance imaging (MRI). Hip-knee-ankle axis (HKAA) and Kellgren-Lawrence (K-L) grades were assessed on simple radiographs. No treatment-related adverse events occurred during the 5-year follow-up. Both VAS and total WOMAC scores improved significantly at 6 months after the injection and until the latest follow-up. Total WORMS was significantly improved until 3 years after the injection. However, the chondral defect size on MRI or other radiologic evaluations did not change significantly. A single intra-articular injection of autologous, high-dose ADMSCs provided safe and clinical improvement without radiologic aggravation for 5 years. Furthermore, structural changes in the osteoarthritic knee showed significant improvement up to 3 years, suggesting a possible option for disease-modifying outpatient treatment for patients with knee OA.
Collapse
Affiliation(s)
- Kang-Il Kim
- Department of Orthopaedic Surgery, Center for Joint Diseases, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
- Department of Orthopaedic Surgery, School of Medicine, Kyung Hee University, Seoul, South Korea
| | - Woo-Suk Lee
- Department of Orthopaedic Surgery, College of Medicine, Gangnam Severance Hospital, Yonsei University, Seoul, South Korea
| | - Jun-Ho Kim
- Corresponding author: Jun-Ho Kim, Department of Orthopaedic Surgery, Center for Joint Diseases, Kyung Hee University Hospital at Gangdong, Seoul, South Korea. Tel: +82-10-7170-0409;
| | - Jung-Kwon Bae
- Department of Orthopaedic Surgery, Center for Joint Diseases, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| | - Wook Jin
- Department of Radiology, Kyung Hee University Hospital at Gandong, Seoul, South Korea
| |
Collapse
|
37
|
Nguyen TPT, Li F, Shrestha S, Tuan RS, Thissen H, Forsythe JS, Frith JE. Cell-laden injectable microgels: Current status and future prospects for cartilage regeneration. Biomaterials 2021; 279:121214. [PMID: 34736147 DOI: 10.1016/j.biomaterials.2021.121214] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/19/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022]
Abstract
Injectable hydrogels have been employed extensively as versatile materials for cartilage regeneration due to their excellent biocompatibility, tunable structure, and ability to accommodate bioactive factors, as well as their ability to be locally delivered via minimally invasive injection to fill irregular defects. More recently, in vitro and in vivo studies have revealed that processing these materials to produce cell-laden microgels can enhance cell-cell and cell-matrix interactions and boost nutrient and metabolite exchange. Moreover, these studies have demonstrated gene expression profiles and matrix regeneration that are superior compared to conventional injectable bulk hydrogels. As cell-laden microgels and their application in cartilage repair are moving closer to clinical translation, this review aims to present an overview of the recent developments in this field. Here we focus on the currently used biomaterials and crosslinking strategies, the innovative fabrication techniques being used for the production of microgels, the cell sources used, the signals used for induction of chondrogenic differentiation and the resultant biological responses, and the ability to create three-dimensional, functional cartilage tissues. In addition, this review also covers the current clinical approaches for repairing cartilage as well as specific challenges faced when attempting the regeneration of damaged cartilage tissue. New findings related to the macroporous nature of the structures formed by the assembled microgel building blocks and the novel use of microgels in 3D printing for cartilage tissue engineering are also highlighted. Finally, we outline the challenges and future opportunities for employing cell-laden microgels in clinical applications.
Collapse
Affiliation(s)
- Thuy P T Nguyen
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Fanyi Li
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Surakshya Shrestha
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Helmut Thissen
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Clayton, VIC 3800, Australia.
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Clayton, VIC 3800, Australia.
| |
Collapse
|
38
|
Kondo M, Kameishi S, Kim K, Metzler NF, Maak TG, Hutchinson DT, Wang AA, Maehara M, Sato M, Grainger DW, Okano T. Safety and efficacy of human juvenile chondrocyte-derived cell sheets for osteochondral defect treatment. NPJ Regen Med 2021; 6:65. [PMID: 34654830 PMCID: PMC8520001 DOI: 10.1038/s41536-021-00173-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/14/2021] [Indexed: 02/06/2023] Open
Abstract
Knee cartilage does not regenerate spontaneously after injury, and a gold standard regenerative treatment algorithm has not been established. This study demonstrates preclinical safety and efficacy of scaffold-free, human juvenile cartilage-derived-chondrocyte (JCC) sheets produced from routine surgical discards using thermo-responsive cultureware. JCCs exhibit stable and high growth potential in vitro over passage 10, supporting possibilities for scale-up to mass production for commercialization. JCC sheets contain highly viable, densely packed cells, show no anchorage-independent cell growth, express mesenchymal surface markers, and lack MHC II expression. In nude rat focal osteochondral defect models, stable neocartilage formation was observed at 4 weeks by JCC sheet transplantation without abnormal tissue growth over 24 weeks in contrast to the nontreatment group showing no spontaneous cartilage repair. Regenerated cartilage was safranin-O positive, contained type II collagen, aggrecan, and human vimentin, and lacked type I collagen, indicating that the hyaline-like neocartilage formed originates from transplanted JCC sheets rather than host-derived cells. This study demonstrates the safety of JCC sheets and stable hyaline cartilage formation with engineered JCC sheets utilizing a sustainable tissue supply. Cost-benefit and scaling issues for sheet fabrication and use support feasibility of this JCC sheet strategy in clinical cartilage repair.
Collapse
Affiliation(s)
- Makoto Kondo
- grid.223827.e0000 0001 2193 0096Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112 USA
| | - Sumako Kameishi
- grid.223827.e0000 0001 2193 0096Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112 USA
| | - Kyungsook Kim
- grid.223827.e0000 0001 2193 0096Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112 USA
| | - Nicolas F. Metzler
- grid.223827.e0000 0001 2193 0096Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112 USA ,grid.223827.e0000 0001 2193 0096Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive SMBB 3100, Salt Lake City, UT 84112 USA
| | - Travis G. Maak
- grid.223827.e0000 0001 2193 0096Department of Orthopaedic Surgery, University of Utah Orthopaedic Center, University of Utah, 590 Wakara Way, Salt Lake City, UT 84108 USA
| | - Douglas T. Hutchinson
- grid.223827.e0000 0001 2193 0096Department of Orthopaedic Surgery, University of Utah Orthopaedic Center, University of Utah, 590 Wakara Way, Salt Lake City, UT 84108 USA ,grid.415178.e0000 0004 0442 6404Pediatric Orthopaedics Surgery, Primary Children’s Hospital Orthopedics, 100 North Mario Capecchi Dr. Suite 4550, Salt Lake City, UT 84113 USA
| | - Angela A. Wang
- grid.223827.e0000 0001 2193 0096Department of Orthopaedic Surgery, University of Utah Orthopaedic Center, University of Utah, 590 Wakara Way, Salt Lake City, UT 84108 USA ,grid.415178.e0000 0004 0442 6404Pediatric Orthopaedics Surgery, Primary Children’s Hospital Orthopedics, 100 North Mario Capecchi Dr. Suite 4550, Salt Lake City, UT 84113 USA
| | - Miki Maehara
- grid.265061.60000 0001 1516 6626Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193 Japan
| | - Masato Sato
- grid.265061.60000 0001 1516 6626Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193 Japan
| | - David W. Grainger
- grid.223827.e0000 0001 2193 0096Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112 USA ,grid.223827.e0000 0001 2193 0096Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive SMBB 3100, Salt Lake City, UT 84112 USA
| | - Teruo Okano
- grid.223827.e0000 0001 2193 0096Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112 USA ,grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan
| |
Collapse
|
39
|
González Vázquez AG, Blokpoel Ferreras LA, Bennett KE, Casey SM, Brama PAJ, O'Brien FJ. Systematic Comparison of Biomaterials-Based Strategies for Osteochondral and Chondral Repair in Large Animal Models. Adv Healthc Mater 2021; 10:e2100878. [PMID: 34405587 PMCID: PMC11468758 DOI: 10.1002/adhm.202100878] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/16/2021] [Indexed: 01/10/2023]
Abstract
Joint repair remains a major challenge in orthopaedics. Recent progress in biomaterial design has led to the fabrication of a plethora of promising devices. Pre-clinical testing of any joint repair strategy typically requires the use of large animal models (e.g., sheep, goat, pig or horse). Despite the key role of such models in clinical translation, there is still a lack of consensus regarding optimal experimental design, making it difficult to draw conclusions on their efficacy. In this context, the authors performed a systematic literature review and a risk of bias assessment on large animal models published between 2010 and 2020, to identify key experimental parameters that significantly affect the biomaterial therapeutic outcome and clinical translation potential (including defect localization, animal age/maturity, selection of controls, cell-free versus cell-laden). They determined that mechanically strong biomaterials perform better at the femoral condyles; while highlighted the importance of including native tissue controls to better evaluate the quality of the newly formed tissue. Finally, in cell-laded biomaterials, the pre-culture conditions played a more important role in defect repair than the cell type. In summary, here they present a systematic evaluation on how the experimental design of preclinical models influences biomaterial-based therapeutic outcomes in joint repair.
Collapse
Affiliation(s)
- Arlyng G. González Vázquez
- Tissue Engineering Research GroupDepartment of Anatomy and Regenerative MedicineRoyal College of Surgeons in Ireland (RCSI)Dublin2 D02 YN77Ireland
- Advanced Materials Bio‐Engineering Research Centre (AMBER)RCSI and TCDDublin2 D02 PN40Ireland
| | - Lia A. Blokpoel Ferreras
- Tissue Engineering Research GroupDepartment of Anatomy and Regenerative MedicineRoyal College of Surgeons in Ireland (RCSI)Dublin2 D02 YN77Ireland
- Advanced Materials Bio‐Engineering Research Centre (AMBER)RCSI and TCDDublin2 D02 PN40Ireland
| | | | - Sarah M. Casey
- Tissue Engineering Research GroupDepartment of Anatomy and Regenerative MedicineRoyal College of Surgeons in Ireland (RCSI)Dublin2 D02 YN77Ireland
- Advanced Materials Bio‐Engineering Research Centre (AMBER)RCSI and TCDDublin2 D02 PN40Ireland
| | - Pieter AJ Brama
- School of Veterinary MedicineUniversity College Dublin (UCD)Dublin4 D04 V1W8Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research GroupDepartment of Anatomy and Regenerative MedicineRoyal College of Surgeons in Ireland (RCSI)Dublin2 D02 YN77Ireland
- Advanced Materials Bio‐Engineering Research Centre (AMBER)RCSI and TCDDublin2 D02 PN40Ireland
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College Dublin (TCD)Dublin2 D02 PN40Ireland
| |
Collapse
|
40
|
Novel therapies using cell sheets engineered from allogeneic mesenchymal stem/stromal cells. Emerg Top Life Sci 2021; 4:677-689. [PMID: 33231260 PMCID: PMC7939697 DOI: 10.1042/etls20200151] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 01/05/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) have long been recognized to help regenerate tissues, by exploiting their intrinsic potentials for differentiation and secretion of therapeutic paracrine factors together with feasibility for cell banking. These unique MSC properties are attractive to provide effective new cell-based therapies for unmet medical needs. Currently, the infusion of suspended MSCs is accepted as a promising therapy to treat systemic inflammatory diseases. However, low cell engraftment/retention in target organs and off-target entrapment using conventional cell infusion must be improved to provide reliable localized disease treatments. Cell sheet technology offers an alternative: three-dimensional (3D) tissue-like structures can be harvested from culture using mild temperature reduction, and transplanted directly onto target tissue sites without suturing, yielding stable cell engraftment and prolonged cell retention in situ without off-target losses. Engineered MSC sheets directly address two major cell therapy strategies based on their therapeutic benefits: (1) tissue replacements based on mult-ilineage differentiation capacities, focusing on cartilage regeneration in this review, and (2) enhancement of tissue recovery via paracrine signaling, employing their various secreted cytokines to promote neovascularization. MSCs also have production benefits as a promising allogeneic cell source by exploiting their reliable proliferative capacity to facilitate expansion and sustainable cell banking for off-the-shelf therapies. This article reviews the advantages of both MSCs as allogeneic cell sources in contrast with autologous cell sources, and allogeneic MSC sheets engineered on thermo-responsive cell dishes as determined in basic studies and clinical achievements, indicating promise to provide robust new cell therapies to future patients.
Collapse
|
41
|
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
|
42
|
García-González J, Marhuenda-Castillo S, Romero-Carretero S, Beltrán-García J. New era of personalized medicine: Advanced therapy medicinal products in Europe. World J Immunol 2021; 11:1-10. [DOI: 10.5411/wji.v11.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
Advanced therapy medicinal products are human medical therapies based on genes, cells, or tissues, and due to their characteristics, they offer new innovative opportunities for the treatment of diseases and injuries, especially for diseases beyond the reach of traditional approaches. These therapies are at the forefront of innovation and have historically been very controversial, although in the last decade they have gained prominence while the number of new advanced therapies has increased every year. In this regard, despite the controversy they may generate, they are expected to dominate the market in the coming decades. Technologies based on advanced therapies are the present and future of medicine and bring us closer to the long-awaited precision medicine. Here we review the field as it stands today, with a focus on the molecular mechanisms that guided the different advanced therapies approved by the European Medicines Agency, their current status, and their legal approval.
Collapse
Affiliation(s)
| | | | | | - Jesús Beltrán-García
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia 46010, Spain
- Center for Biomedical Research in Rare Diseases Network (CIBERER), Carlos III Health Institute, Valencia 46010, Spain
- INCLIVA Institute of Sanitary Research, Valencia 46010, Spain
| |
Collapse
|
43
|
Pei YA, Dong Y, He TC, Li WJ, Toh WS, Pei M. Editorial: Extracellular Vesicle Treatment, Epigenetic Modification and Cell Reprogramming to Promote Bone and Cartilage Regeneration. Front Bioeng Biotechnol 2021; 9:678014. [PMID: 33968918 PMCID: PMC8096898 DOI: 10.3389/fbioe.2021.678014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Yufeng Dong
- Department of Orthopedic Surgery, Center for Tissue Engineering and Regenerative Medicine, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Wan-Ju Li
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, United States
| | - Wei Seong Toh
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
44
|
Thorp H, Kim K, Kondo M, Maak T, Grainger DW, Okano T. Trends in Articular Cartilage Tissue Engineering: 3D Mesenchymal Stem Cell Sheets as Candidates for Engineered Hyaline-Like Cartilage. Cells 2021; 10:cells10030643. [PMID: 33805764 PMCID: PMC7998529 DOI: 10.3390/cells10030643] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Articular cartilage defects represent an inciting factor for future osteoarthritis (OA) and degenerative joint disease progression. Despite multiple clinically available therapies that succeed in providing short term pain reduction and restoration of limited mobility, current treatments do not reliably regenerate native hyaline cartilage or halt cartilage degeneration at these defect sites. Novel therapeutics aimed at addressing limitations of current clinical cartilage regeneration therapies increasingly focus on allogeneic cells, specifically mesenchymal stem cells (MSCs), as potent, banked, and available cell sources that express chondrogenic lineage commitment capabilities. Innovative tissue engineering approaches employing allogeneic MSCs aim to develop three-dimensional (3D), chondrogenically differentiated constructs for direct and immediate replacement of hyaline cartilage, improve local site tissue integration, and optimize treatment outcomes. Among emerging tissue engineering technologies, advancements in cell sheet tissue engineering offer promising capabilities for achieving both in vitro hyaline-like differentiation and effective transplantation, based on controlled 3D cellular interactions and retained cellular adhesion molecules. This review focuses on 3D MSC-based tissue engineering approaches for fabricating “ready-to-use” hyaline-like cartilage constructs for future rapid in vivo regenerative cartilage therapies. We highlight current approaches and future directions regarding development of MSC-derived cartilage therapies, emphasizing cell sheet tissue engineering, with specific focus on regulating 3D cellular interactions for controlled chondrogenic differentiation and post-differentiation transplantation capabilities.
Collapse
Affiliation(s)
- Hallie Thorp
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Department of Biomedical Engineering, University of Utah, 36 S Wasatch Dr, Salt Lake City, UT 84112, USA
| | - Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Correspondence: (K.K.); (T.O.); Tel.: +1-801-585-0070 (K.K. & T.O.); Fax: +1-801-581-3674 (K.K. & T.O.)
| | - Makoto Kondo
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
| | - Travis Maak
- Department of Orthopaedic Surgery, University of Utah, 590 Wakara Way, Salt Lake City, UT 84108, USA;
| | - David W. Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Department of Biomedical Engineering, University of Utah, 36 S Wasatch Dr, Salt Lake City, UT 84112, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Wakamatsucho, 2−2, Shinjuku-ku, Tokyo 162-8480, Japan
- Correspondence: (K.K.); (T.O.); Tel.: +1-801-585-0070 (K.K. & T.O.); Fax: +1-801-581-3674 (K.K. & T.O.)
| |
Collapse
|
45
|
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
|
46
|
Futrega K, Music E, Robey PG, Gronthos S, Crawford R, Saifzadeh S, Klein TJ, Doran MR. Characterisation of ovine bone marrow-derived stromal cells (oBMSC) and evaluation of chondrogenically induced micro-pellets for cartilage tissue repair in vivo. Stem Cell Res Ther 2021; 12:26. [PMID: 33413652 PMCID: PMC7791713 DOI: 10.1186/s13287-020-02045-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract Bone marrow stromal cells (BMSC) show promise in cartilage repair, and sheep are the most common large animal pre-clinical model. Objective The objective of this study was to characterise ovine BMSC (oBMSC) in vitro, and to evaluate the capacity of chondrogenic micro-pellets manufactured from oBMSC or ovine articular chondrocytes (oACh) to repair osteochondral defects in sheep. Design oBMSC were characterised for surface marker expression using flow cytometry and evaluated for tri-lineage differentiation capacity. oBMSC micro-pellets were manufactured in a microwell platform, and chondrogenesis was compared at 2%, 5%, and 20% O2. The capacity of cartilage micro-pellets manufactured from oBMSC or oACh to repair osteochondral defects in adult sheep was evaluated in an 8-week pilot study. Results Expanded oBMSC were positive for CD44 and CD146 and negative for CD45. The common adipogenic induction ingredient, 3-Isobutyl-1-methylxanthine (IBMX), was toxic to oBMSC, but adipogenesis could be restored by excluding IBMX from the medium. BMSC chondrogenesis was optimal in a 2% O2 atmosphere. Micro-pellets formed from oBMSC or oACh appeared morphologically similar, but hypertrophic genes were elevated in oBMSC micro-pellets. While oACh micro-pellets formed cartilage-like repair tissue in sheep, oBMSC micro-pellets did not. Conclusion The sensitivity of oBMSC, compared to human BMSC, to IBMX in standard adipogenic assays highlights species-associated differences. Micro-pellets manufactured from oACh were more effective than micro-pellets manufactured from oBMSC in the repair of osteochondral defects in sheep. While oBMSC can be driven to form cartilage-like tissue in vitro, the effective use of these cells in cartilage repair will depend on the successful mitigation of hypertrophy and tissue integration. Supplementary information The online version contains supplementary material available at 10.1186/s13287-020-02045-3.
Collapse
Affiliation(s)
- K Futrega
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia.,National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA.,Translational Research Institute (TRI), Brisbane, Queensland, Australia
| | - E Music
- Translational Research Institute (TRI), Brisbane, Queensland, Australia.,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - P G Robey
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - S Gronthos
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - R Crawford
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - S Saifzadeh
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - T J Klein
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - M R Doran
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia. .,National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA. .,Translational Research Institute (TRI), Brisbane, Queensland, Australia. .,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia. .,Mater Research Institute - University of Queensland (UQ), Translational Research Institute (TRI), Brisbane, Queensland, Australia.
| |
Collapse
|
47
|
Futrega K, Robey PG, Klein TJ, Crawford RW, Doran MR. A single day of TGF-β1 exposure activates chondrogenic and hypertrophic differentiation pathways in bone marrow-derived stromal cells. Commun Biol 2021; 4:29. [PMID: 33398032 PMCID: PMC7782775 DOI: 10.1038/s42003-020-01520-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 11/24/2020] [Indexed: 01/29/2023] Open
Abstract
Virtually all bone marrow-derived stromal cell (BMSC) chondrogenic induction cultures include greater than 2 weeks exposure to transforming growth factor-β (TGF-β), but fail to generate cartilage-like tissue suitable for joint repair. Herein we used a micro-pellet model (5 × 103 BMSC each) to determine the duration of TGF-β1 exposure required to initiate differentiation machinery, and to characterize the role of intrinsic programming. We found that a single day of TGF-β1 exposure was sufficient to trigger BMSC chondrogenic differentiation and tissue formation, similar to 21 days of TGF-β1 exposure. Despite cessation of TGF-β1 exposure following 24 hours, intrinsic programming mediated further chondrogenic and hypertrophic BMSC differentiation. These important behaviors are obfuscated by diffusion gradients and heterogeneity in commonly used macro-pellet models (2 × 105 BMSC each). Use of more homogenous micro-pellet models will enable identification of the critical differentiation cues required, likely in the first 24-hours, to generate high quality cartilage-like tissue from BMSC.
Collapse
Affiliation(s)
- Kathryn Futrega
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD, USA
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
- Translational Research Institute (TRI), Brisbane, Queensland, Australia
| | - Pamela G Robey
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD, USA
| | - Travis J Klein
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Ross W Crawford
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Michael R Doran
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD, USA.
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia.
- Translational Research Institute (TRI), Brisbane, Queensland, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, Australia.
- Mater Research Institute, University of Queensland (UQ), Brisbane, Queensland, Australia.
| |
Collapse
|
48
|
Taghiyar L, Jahangir S, Khozaei Ravari M, Shamekhi MA, Eslaminejad MB. Cartilage Repair by Mesenchymal Stem Cell-Derived Exosomes: Preclinical and Clinical Trial Update and Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1326:73-93. [PMID: 33629260 DOI: 10.1007/5584_2021_625] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) and other degenerative joint diseases are characterized by articular cartilage destruction, synovial inflammation, sclerosis of subchondral bone, and loss of extracellular matrix (ECM). Worldwide, these diseases are major causes of disability. Cell therapies have been considered to be the best therapeutic strategies for long-term treatment of articular cartilage diseases. It has been suggested that the mechanism of stem cell-based therapy is related to paracrine secretion of extracellular vesicles (EVs), which are recognized as the main secretion factors of stem cells. EVs, and in particular the subclass exosomes (Exos), are novel therapeutic approaches for treatment of cartilage lesions and OA. The results of recent studies have shown that EVs isolated from mesenchymal stem cells (MSCs) could inhibit OA progression. EVs isolated from various stem cell sources, such as MSCs, may contribute to tissue regeneration of the limbs, skin, heart, and other tissues. Here, we summarize recent findings of preclinical and clinical studies on different MSC-derived EVs and their effectiveness as a treatment for damaged cartilage. The Exos isolation techniques in OA treatment are also highlighted.
Collapse
Affiliation(s)
- Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahrbano Jahangir
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mojtaba Khozaei Ravari
- 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.
| |
Collapse
|
49
|
Sekar MP, Budharaju H, Zennifer A, Sethuraman S, Vermeulen N, Sundaramurthi D, Kalaskar DM. Current standards and ethical landscape of engineered tissues-3D bioprinting perspective. J Tissue Eng 2021; 12:20417314211027677. [PMID: 34377431 PMCID: PMC8330463 DOI: 10.1177/20417314211027677] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/08/2021] [Indexed: 01/17/2023] Open
Abstract
Tissue engineering is an evolving multi-disciplinary field with cutting-edge technologies and innovative scientific perceptions that promise functional regeneration of damaged tissues/organs. Tissue engineered medical products (TEMPs) are biomaterial-cell products or a cell-drug combination which is injected, implanted or topically applied in the course of a therapeutic or diagnostic procedure. Current tissue engineering strategies aim at 3D printing/bioprinting that uses cells and polymers to construct living tissues/organs in a layer-by-layer fashion with high 3D precision. However, unlike conventional drugs or therapeutics, TEMPs and 3D bioprinted tissues are novel therapeutics and need different regulatory protocols for clinical trials and commercialization processes. Therefore, it is essential to understand the complexity of raw materials, cellular components, and manufacturing procedures to establish standards that can help to translate these products from bench to bedside. These complexities are reflected in the regulations and standards that are globally in practice to prevent any compromise or undue risks to patients. This review comprehensively describes the current legislations, standards for TEMPs with a special emphasis on 3D bioprinted tissues. Based on these overviews, challenges in the clinical translation of TEMPs & 3D bioprinted tissues/organs along with their ethical concerns and future perspectives are discussed.
Collapse
Affiliation(s)
- Muthu Parkkavi Sekar
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Allen Zennifer
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Niki Vermeulen
- Department of Science, Technology and Innovation Studies, School of Social and Political Science, University of Edinburgh, High School Yards, Edinburgh, UK
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | | |
Collapse
|
50
|
Kim JK, Bae HC, Ro DH, Lee S, Lee MC, Han HS. Enhancement of Cartilage Regeneration of Synovial Stem Cells/Hydrogel by Using Transglutaminase-4. Tissue Eng Part A 2020; 27:761-770. [PMID: 33107390 DOI: 10.1089/ten.tea.2020.0271] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although mesenchymal stem cells (MSCs) transplantation is reportedly a promising strategy for repairing damaged articular cartilage, MSCs-based cartilage tissue engineering has numerous limitations, including poor implanted cell adhesion, phenotypic alteration of cells, regulation of mechanical properties, and engraftment rates after implantation. This study aimed to investigate the efficacy of transplantation of synovium-derived mesenchymal stem cells (SDSCs) encapsulated in a hyaluronic acid/collagen/fibrinogen (HA/COL/FG) composite gel by supplementing recombinant human transglutaminase 4 (rhTG-4) in treating osteochondral defects. RhTG-4 treatment induced the expression of integrin β1 and dynamic actin fiber, enhancing SDSCs adhesion to fibronectin. Supplementation of rhTG-4 significantly induced the proliferation of SDSCs encapsulated in the HA/COL/FG composite gel and increased the hardness of the extracellular matrix. Furthermore, supplementation of rhTG-4 significantly upregulated aggrecan and type II collagen mRNA. Pretreatment with integrin β1 siRNA markedly suppressed TG4-induced actin remodeling, activation mitogen-activated protein kinase (MAPK), and eventually the chondrogenesis-related genes. Moreover, transplantation of SDSCs encapsulated in HA/COL/FG/rhTG-4 composite gel in vivo yielded reconstructed tissue resembling native hyaline cartilage. These data suggest that rhTG-4 enhances cartilage regeneration of the SDSCs encapsulated in hydrogel in rabbits. Impact statement In this study, we investigated the effects of recombinant human transglutaminase 4 on the ability of synovium-derived mesenchymal stem cells encapsulated in a hyaluronic acid/collagen/fibrinogen composite gel to repair osteochondral defects. We believe that our study makes a significant contribution to the literature because it explores a method of improving an existing modality to mediate tissue repair.
Collapse
Affiliation(s)
- Jong-Keun Kim
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Cheol Bae
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Du Hyun Ro
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sahnghoon Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Myung Chul Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyuk-Soo Han
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|