Cell Therapies in Orthopaedics

A special issue of Cells (ISSN 2073-4409).

Deadline for manuscript submissions: closed (28 February 2022) | Viewed by 41957

Special Issue Editors


E-Mail Website
Guest Editor
School of Pharmacy and Bioengineering, Keele University (UK) based at the RJAH Orthopaedic Hospital, Oswestry, UK
Interests: regenerative medicine; orthopaedics; Spinal cord injury; pressure ulcers; biomarkers; prognostic/predictive modelling

E-Mail Website
Guest Editor
School of Pharmacy and Bioengineering, Keele University (UK) based at the RJAH Orthopaedic Hospital, Oswestry, UK
Interests: orthopaedics; regenerative medicine; biomarkers; cell therapy; bioinformatics; proteomics; spinal cord injury; prognostic/predictive modelling

Special Issue Information

Dear Colleagues,

Treating neuromusculoskeletal disorders successfully using cell-based therapies and preventing or delaying the debilitating symptoms of chronic/inflammatory conditions such as osteoarthritis and rheumatoid arthritis would have a huge impact on human health in an aging worldwide population. Cell-based therapies in the orthopaedic clinical setting have existed for around 30 years and currently there are several in use or in clinical trials around the globe for the treatment of neuromusculoskeletal disorders. An improved understanding of the precise mechanisms of action for these cell-based therapies and the discovery and validation of predictive clinical indicators (including biomarkers) are priorities for the widespread translation and global application of burgeoning orthopaedic cell-based therapies.

The aim of this Special Issue is to provide the current ‘state-of-play’ in the orthopaedic cell therapy field, with a particular focus on mechanisms of action, tissue engineering approaches, in vitro systems, preclinical models, clinical trials, and predictive clinical indicators (including biomarkers), highlighting any barriers that have been identified that might hinder the translation or widespread adoption of these techniques.

Dr. Karina Theresa Wright
Dr. Charlotte Hulme
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • cell therapy
  • tissue engineering
  • in vitro models
  • preclinical models
  • clinical trials
  • precision medicine
  • biomarkers
  • neuromusculoskeletal disorders
  • orthopaedics
  • arthritis
  • spinal disorders

Published Papers (13 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

30 pages, 4374 KiB  
Article
Biodegradable Poly(D-L-lactide-co-glycolide) (PLGA)-Infiltrated Bioactive Glass (CAR12N) Scaffolds Maintain Mesenchymal Stem Cell Chondrogenesis for Cartilage Tissue Engineering
by Clemens Gögele, Silvana Müller, Svetlana Belov, Andreas Pradel, Sven Wiltzsch, Armin Lenhart, Markus Hornfeck, Vera Kerling, Achim Rübling, Hannes Kühl, Kerstin Schäfer-Eckart, Bernd Minnich, Thomas Martin Weiger and Gundula Schulze-Tanzil
Cells 2022, 11(9), 1577; https://doi.org/10.3390/cells11091577 - 7 May 2022
Cited by 5 | Viewed by 2589
Abstract
Regeneration of articular cartilage remains challenging. The aim of this study was to increase the stability of pure bioactive glass (BG) scaffolds by means of solvent phase polymer infiltration and to maintain cell adherence on the glass struts. Therefore, BG scaffolds either pure [...] Read more.
Regeneration of articular cartilage remains challenging. The aim of this study was to increase the stability of pure bioactive glass (BG) scaffolds by means of solvent phase polymer infiltration and to maintain cell adherence on the glass struts. Therefore, BG scaffolds either pure or enhanced with three different amounts of poly(D-L-lactide-co-glycolide) (PLGA) were characterized in detail. Scaffolds were seeded with primary porcine articular chondrocytes (pACs) and human mesenchymal stem cells (hMSCs) in a dynamic long-term culture (35 days). Light microscopy evaluations showed that PLGA was detectable in every region of the scaffold. Porosity was greater than 70%. The biomechanical stability was increased by polymer infiltration. PLGA infiltration did not result in a decrease in viability of both cell types, but increased DNA and sulfated glycosaminoglycan (sGAG) contents of hMSCs-colonized scaffolds. Successful chondrogenesis of hMSC-colonized scaffolds was demonstrated by immunocytochemical staining of collagen type II, cartilage proteoglycans and the transcription factor SOX9. PLGA-infiltrated scaffolds showed a higher relative expression of cartilage related genes not only of pAC-, but also of hMSC-colonized scaffolds in comparison to the pure BG. Based on the novel data, our recommendation is BG scaffolds with single infiltrated PLGA for cartilage tissue engineering. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

36 pages, 6963 KiB  
Article
Retrospective Analysis of Autologous Chondrocyte-Based Cytotherapy Production for Clinical Use: GMP Process-Based Manufacturing Optimization in a Swiss University Hospital
by Virginie Philippe, Alexis Laurent, Nathalie Hirt-Burri, Philippe Abdel-Sayed, Corinne Scaletta, Valentine Schneebeli, Murielle Michetti, Jean-François Brunet, Lee Ann Applegate and Robin Martin
Cells 2022, 11(6), 1016; https://doi.org/10.3390/cells11061016 - 17 Mar 2022
Cited by 8 | Viewed by 2790
Abstract
Cultured autologous human articular chondrocyte (HAC) implantation has been extensively investigated for safe and effective promotion of structural and functional restoration of knee cartilage lesions. HAC-based cytotherapeutic products for clinical use must be manufactured under an appropriate quality assurance system and follow good [...] Read more.
Cultured autologous human articular chondrocyte (HAC) implantation has been extensively investigated for safe and effective promotion of structural and functional restoration of knee cartilage lesions. HAC-based cytotherapeutic products for clinical use must be manufactured under an appropriate quality assurance system and follow good manufacturing practices (GMP). A prospective clinical trial is ongoing in the Lausanne University Hospital, where the HAC manufacturing processes have been implemented internally. Following laboratory development and in-house GMP transposition of HAC cell therapy manufacturing, a total of 47 patients have been treated to date. The main aim of the present study was to retrospectively analyze the available manufacturing records of the produced HAC-based cytotherapeutic products, outlining the inter-individual variability existing among the 47 patients regarding standardized transplant product preparation. These data were used to ameliorate and to ensure the continued high quality of cytotherapeutic care in view of further clinical investigations, based on the synthetic analyses of existing GMP records. Therefore, a renewed risk analysis-based process definition was performed, with specific focus set on process parameters, controls, targets, and acceptance criteria. Overall, high importance of the interdisciplinary collaboration and of the manufacturing process robustness was underlined, considering the high variability (i.e., quantitative, functional) existing between the treated patients and between the derived primary HAC cell types. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

18 pages, 2480 KiB  
Article
Metabolomic Profiling and Mechanotransduction of Single Chondrocytes Encapsulated in Alginate Microgels
by Jacob P. Fredrikson, Priyanka P. Brahmachary, Ayten E. Erdoğan, Zachary K. Archambault, James N. Wilking, Ronald K. June and Connie B. Chang
Cells 2022, 11(5), 900; https://doi.org/10.3390/cells11050900 - 5 Mar 2022
Cited by 8 | Viewed by 3064
Abstract
Articular cartilage is comprised of two main components, the extracellular matrix (ECM) and the pericellular matrix (PCM). The PCM helps to protect chondrocytes in the cartilage from mechanical loads, but in patients with osteoarthritis, the PCM is weakened, resulting in increased chondrocyte stress. [...] Read more.
Articular cartilage is comprised of two main components, the extracellular matrix (ECM) and the pericellular matrix (PCM). The PCM helps to protect chondrocytes in the cartilage from mechanical loads, but in patients with osteoarthritis, the PCM is weakened, resulting in increased chondrocyte stress. As chondrocytes are responsible for matrix synthesis and maintenance, it is important to understand how mechanical loads affect the cellular responses of chondrocytes. Many studies have examined chondrocyte responses to in vitro mechanical loading by embedding chondrocytes in 3-D hydrogels. However, these experiments are mostly performed in the absence of PCM, which may obscure important responses to mechanotransduction. Here, drop-based microfluidics is used to culture single chondrocytes in alginate microgels for cell-directed PCM synthesis that closely mimics the in vivo microenvironment. Chondrocytes formed PCM over 10 days in these single-cell 3-D microenvironments. Mechanotransduction studies were performed, in which single-cell microgels mimicking the cartilage PCM were embedded in high-stiffness agarose. After physiological dynamic compression in a custom-built bioreactor, microgels exhibited distinct metabolomic profiles from both uncompressed and monolayer controls. These results demonstrate the potential of single cell encapsulation in alginate microgels to advance cartilage tissue engineering and basic chondrocyte mechanobiology. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

17 pages, 967 KiB  
Article
Osteochondral Lesions of the Ankle Treated with Bone Marrow Concentrate with Hyaluronan and Fibrin: A Single-Centre Study
by Sameera Abas, Jan Herman Kuiper, Sally Roberts, Helen McCarthy, Mike Williams, Andrew Bing, Bernhard Tins and Nilesh Makwana
Cells 2022, 11(4), 629; https://doi.org/10.3390/cells11040629 - 11 Feb 2022
Cited by 4 | Viewed by 2299
Abstract
Osteochondral defects of the ankle (OCD) are being increasingly identified as a clinically significant consequence of injury to the ankle, with the potential to lead to osteoarthritis if left untreated. The aim of this retrospective cohort study was to evaluate a single-stage treatment [...] Read more.
Osteochondral defects of the ankle (OCD) are being increasingly identified as a clinically significant consequence of injury to the ankle, with the potential to lead to osteoarthritis if left untreated. The aim of this retrospective cohort study was to evaluate a single-stage treatment of OCD, based on bone marrow aspirate (BMA) centrifuged to produce bone marrow concentrate (BMC). In a dual syringe, the concentrate was mixed with thrombin in one syringe, whereas hyaluronan and fibrinogen were mixed in a second syringe. The two mixtures were then injected and combined into the prepared defect. Clinical outcome and quality of life scores (MOXFQ and EQ-5D) were collected at baseline and yearly thereafter. Multilevel models were used to analyse the pattern of scores over time. Ninety-four patients were treated between 2015 and 2020. The means of each of the three components of the MOXFQ significantly improved between baseline and 1 year (p < 0.001 for each component), with no further change from year 1 to year 3. The EQ-5D index also improved significantly from baseline to 1 year, with no evidence for further change. Our results strongly indicate that this BMC treatment is safe for, and well tolerated by, patients with OCD of the ankle as both primary treatment and those who have failed primary treatment. This technique provides a safe, efficacious alternative to currently employed cartilage repair techniques, with favourable outcomes and a low complication rate at 36 months. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

17 pages, 3057 KiB  
Article
Microvalve Bioprinting of MSC-Chondrocyte Co-Cultures
by Joseph Dudman, Ana Marina Ferreira, Piergiorgio Gentile, Xiao Wang and Kenneth Dalgarno
Cells 2021, 10(12), 3329; https://doi.org/10.3390/cells10123329 - 27 Nov 2021
Cited by 2 | Viewed by 2260
Abstract
Recent improvements within the fields of high-throughput screening and 3D tissue culture have provided the possibility of developing in vitro micro-tissue models that can be used to study diseases and screen potential new therapies. This paper reports a proof-of-concept study on the use [...] Read more.
Recent improvements within the fields of high-throughput screening and 3D tissue culture have provided the possibility of developing in vitro micro-tissue models that can be used to study diseases and screen potential new therapies. This paper reports a proof-of-concept study on the use of microvalve-based bioprinting to create laminar MSC-chondrocyte co-cultures to investigate whether the use of MSCs in ACI procedures would stimulate enhanced ECM production by chondrocytes. Microvalve-based bioprinting uses small-scale solenoid valves (microvalves) to deposit cells suspended in media in a consistent and repeatable manner. In this case, MSCs and chondrocytes have been sequentially printed into an insert-based transwell system in order to create a laminar co-culture, with variations in the ratios of the cell types used to investigate the potential for MSCs to stimulate ECM production. Histological and indirect immunofluorescence staining revealed the formation of dense tissue structures within the chondrocyte and MSC-chondrocyte cell co-cultures, alongside the establishment of a proliferative region at the base of the tissue. No stimulatory or inhibitory effect in terms of ECM production was observed through the introduction of MSCs, although the potential for an immunomodulatory benefit remains. This study, therefore, provides a novel method to enable the scalable production of therapeutically relevant micro-tissue models that can be used for in vitro research to optimise ACI procedures. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

19 pages, 4864 KiB  
Article
Mesenchymal Stromal Cell Differentiation for Generating Cartilage and Bone-Like Tissues In Vitro
by Graziana Monaco, Yann D. Ladner, Alicia J. El Haj, Nicholas R. Forsyth, Mauro Alini and Martin J. Stoddart
Cells 2021, 10(8), 2165; https://doi.org/10.3390/cells10082165 - 22 Aug 2021
Cited by 4 | Viewed by 3575
Abstract
In the field of tissue engineering, progress has been made towards the development of new treatments for cartilage and bone defects. However, in vitro culture conditions for human bone marrow mesenchymal stromal cells (hBMSCs) have not yet been fully defined. To improve our [...] Read more.
In the field of tissue engineering, progress has been made towards the development of new treatments for cartilage and bone defects. However, in vitro culture conditions for human bone marrow mesenchymal stromal cells (hBMSCs) have not yet been fully defined. To improve our understanding of cartilage and bone in vitro differentiation, we investigated the effect of culture conditions on hBMSC differentiation. We hypothesized that the use of two different culture media including specific growth factors, TGFβ1 or BMP2, as well as low (2% O2) or high (20% O2) oxygen tension, would improve the chondrogenic and osteogenic potential, respectively. Chondrogenic and osteogenic differentiation of hBMSCs isolated from multiple donors and expanded under the same conditions were directly compared. Chondrogenic groups showed a notable upregulation of chondrogenic markers compared with osteogenic groups. Greater sGAG production and deposition, and collagen type II and I accumulation occurred for chondrogenic groups. Chondrogenesis at 2% O2 significantly reduced ALP gene expression and reduced type I collagen deposition, producing a more stable and less hypertrophic chondrogenic phenotype. An O2 tension of 2% did not inhibit osteogenic differentiation at the protein level but reduced ALP and OC gene expression. An upregulation of ALP and OC occurred during osteogenesis in BMP2 containing media under 20% O2; BMP2 free osteogenic media downregulated ALP and also led to higher sGAG release. A higher mineralization was observed in the presence of BMP2 during osteogenesis. This study demonstrates how the modulation of O2 tension, combined with tissue-specific growth factors and media composition can be tailored in vitro to promote chondral or endochondral differentiation while using the same donor cell population. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

16 pages, 2598 KiB  
Article
New Insights into Xenotransplantation for Cartilage Repair: Porcine Multi-Genetically Modified Chondrocytes as a Promising Cell Source
by Hanna Tritschler, Konrad Fischer, Jochen Seissler, Jörg Fiedler, Rebecca Halbgebauer, Markus Huber-Lang, Angelika Schnieke and Rolf E. Brenner
Cells 2021, 10(8), 2152; https://doi.org/10.3390/cells10082152 - 20 Aug 2021
Cited by 7 | Viewed by 2851
Abstract
Transplantation of xenogenic porcine chondrocytes could represent a future strategy for the treatment of human articular cartilage defects. Major obstacles are humoral and cellular rejection processes triggered by xenogenic epitopes like α-1,3-Gal and Neu5Gc. Besides knockout (KO) of genes responsible for the biosynthesis [...] Read more.
Transplantation of xenogenic porcine chondrocytes could represent a future strategy for the treatment of human articular cartilage defects. Major obstacles are humoral and cellular rejection processes triggered by xenogenic epitopes like α-1,3-Gal and Neu5Gc. Besides knockout (KO) of genes responsible for the biosynthesis of respective epitopes (GGTA1 and CMAH), transgenic expression of human complement inhibitors and anti-apoptotic as well as anti-inflammatory factors (CD46, CD55, CD59, TNFAIP3 and HMOX1) could synergistically prevent hyperacute xenograft rejection. Therefore, chondrocytes from different strains of single- or multi-genetically modified pigs were characterized concerning their protection from xenogeneic complement activation. Articular chondrocytes were isolated from the knee joints of WT, GalTKO, GalT/CMAH-KO, human CD59/CD55//CD46/TNFAIP3/HMOX1-transgenic (TG), GalTKO/TG and GalT/CMAHKO/TG pigs. The tissue-specific effectiveness of the genetic modifications was tested on gene, protein and epitope expression level or by functional assays. After exposure to 20% and 40% normal human serum (NHS), deposition of C3b/iC3b/C3c and formation of the terminal complement complex (TCC, C5b-9) was quantified by specific cell ELISAs, and generation of the anaphylatoxin C5a by ELISA. Chondrocyte lysis was analyzed by Trypan Blue Exclusion Assay. In all respective KO variants, the absence of α -1,3-Gal and Neu5Gc epitope was verified by FACS analysis. In chondrocytes derived from TG animals, expression of CD55 and CD59 could be confirmed on gene and protein level, TNFAIP3 on gene expression level as well as by functional assays and CD46 only on gene expression level whereas transgenic HMOX1 expression was not evident. Complement activation in the presence of NHS indicated mainly effective although incomplete protection against C3b/iC3b/C3c deposition, C5a-generation and C5b-9 formation being lowest in single GalTKO. Chondrocyte viability under exposure to NHS was significantly improved even by single GalTKO and completely preserved by all other variants including TG chondrocytes without KO of xenoepitopes. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

16 pages, 4104 KiB  
Article
Human Mesenchymal Stromal Cells Enhance Cartilage Healing in a Murine Joint Surface Injury Model
by Jade Perry, Anke J. Roelofs, Claire Mennan, Helen S. McCarthy, Alison Richmond, Susan M. Clark, Anna H. K. Riemen, Karina Wright, Cosimo De Bari and Sally Roberts
Cells 2021, 10(8), 1999; https://doi.org/10.3390/cells10081999 - 6 Aug 2021
Cited by 6 | Viewed by 2720
Abstract
Human umbilical cord (hUC)- or bone marrow (hBM)-derived mesenchymal stromal cells (MSCs) were evaluated as an allogeneic source of cells for cartilage repair. We aimed to determine if they could enhance healing of chondral defects with or without the recruitment of endogenous cells. [...] Read more.
Human umbilical cord (hUC)- or bone marrow (hBM)-derived mesenchymal stromal cells (MSCs) were evaluated as an allogeneic source of cells for cartilage repair. We aimed to determine if they could enhance healing of chondral defects with or without the recruitment of endogenous cells. hMSCs were applied into a focal joint surface injury in knees of adult mice expressing tdTomato fluorescent protein in cells descending from Gdf5-expressing embryonic joint interzone cells. Three experimental groups were used: (i) hUC-MSCs, (ii) hBM-MSCs and (iii) PBS (vehicle) without cells. Cartilage repair was assessed after 8 weeks and tdTomato-expressing cells were detected by immunostaining. Plasma levels of pro-inflammatory mediators and other markers were measured by electrochemiluminescence. Both hUC-MSC (n = 14, p = 0.009) and hBM-MSC (n = 13, p = 0.006) treatment groups had significantly improved cartilage repair compared to controls (n = 18). While hMSCs were not detectable in the repair tissue at 8 weeks post-implantation, increased endogenous Gdf5-lineage cells were detected in repair tissue of hUC-MSC-treated mice. This xenogeneic study indicates that hMSCs enhance intrinsic cartilage repair mechanisms in mice. Hence, hMSCs, particularly the more proliferative hUC-MSCs, could represent an attractive allogeneic cell population for treating patients with chondral defects and perhaps prevent the onset and progression of osteoarthritis. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

23 pages, 12785 KiB  
Article
An In Vitro System to Study the Effect of Subchondral Bone Health on Articular Cartilage Repair in Humans
by Timothy Hopkins, Karina T. Wright, Nicola J. Kuiper, Sally Roberts, Paul Jermin, Peter Gallacher and Jan Herman Kuiper
Cells 2021, 10(8), 1903; https://doi.org/10.3390/cells10081903 - 27 Jul 2021
Cited by 3 | Viewed by 2526
Abstract
Chondrocyte-based cartilage repair strategies, such as articular chondrocyte implantation, are widely used, but few studies addressed the communication between native subchondral bone cells and the transplanted chondrocytes. An indirect co-culture model was developed, representing a chondrocyte/scaffold-construct repair of a cartilage defect adjoining bone, [...] Read more.
Chondrocyte-based cartilage repair strategies, such as articular chondrocyte implantation, are widely used, but few studies addressed the communication between native subchondral bone cells and the transplanted chondrocytes. An indirect co-culture model was developed, representing a chondrocyte/scaffold-construct repair of a cartilage defect adjoining bone, where the bone could have varying degrees of degeneration. Human BM-MSCs were isolated from two areas of subchondral bone in each of five osteochondral tissue specimens from five patients undergoing knee arthroplasty. These two areas underlaid the macroscopically and histologically best and worst cartilage, representing early and late-stage OA, respectively. BM-MSCs were co-cultured with normal chondrocytes suspended in agarose, with the two cell types separated by a porous membrane. After 0, 7, 14 and 21 days, chondrocyte–agarose scaffolds were assessed by gene expression and biochemical analyses, and the abundance of selected proteins in conditioned media was assessed by ELISA. Co-culture with late-OA BM-MSCs resulted in a reduction in GAG deposition and a decreased expression of genes encoding matrix-specific proteins (COL2A1 and ACAN), compared to culturing with early OA BM-MSCs. The concentration of TGF-β1 was significantly higher in the early OA conditioned media. The results of this study have clinical implications for cartilage repair, suggesting that the health of the subchondral bone may influence the outcomes of chondrocyte-based repair strategies. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

16 pages, 4930 KiB  
Article
Short-Term Evaluation of Cellular Fate in an Ovine Bone Formation Model
by Hareklea Markides, Nicola C. Foster, Jane S. McLaren, Timothy Hopkins, Cameron Black, Richard O. C. Oreffo, Brigitte E. Scammell, Iria Echevarria, Lisa J. White and Alicia J. El Haj
Cells 2021, 10(7), 1776; https://doi.org/10.3390/cells10071776 - 14 Jul 2021
Cited by 3 | Viewed by 2749
Abstract
The ovine critical-sized defect model provides a robust preclinical model for testing tissue-engineered constructs for use in the treatment of non-union bone fractures and severe trauma. A critical question in cell-based therapies is understanding the optimal therapeutic cell dose. Key to defining the [...] Read more.
The ovine critical-sized defect model provides a robust preclinical model for testing tissue-engineered constructs for use in the treatment of non-union bone fractures and severe trauma. A critical question in cell-based therapies is understanding the optimal therapeutic cell dose. Key to defining the dose and ensuring successful outcomes is understanding the fate of implanted cells, e.g., viability, bio-distribution and exogenous infiltration post-implantation. This study evaluates such parameters in an ovine critical-sized defect model 2 and 7 days post-implantation. The fate of cell dose and behaviour post-implantation when combined with nanomedicine approaches for multi-model tracking and remote control using external magnetic fields is also addressed. Autologous STRO-4 selected mesenchymal stromal cells (MSCs) were labelled with a fluorescent lipophilic dye (CM-Dil), functionalised magnetic nanoparticles (MNPs) and delivered to the site within a naturally derived bone extracellular matrix (ECM) gel. Encapsulated cells were implanted within a critical-sized defect in an ovine medial femoral condyle and exposed to dynamic gradients of external magnetic fields for 1 h per day. Sheep were sacrificed at 2 and 7 days post-initial surgery where ECM was harvested. STRO-4-positive (STRO-4+) stromal cells expressed osteocalcin and survived within the harvested gels at day 2 and day 7 with a 50% loss at day 2 and a further 45% loss at 7 days. CD45-positive leucocytes were also observed in addition to endogenous stromal cells. No elevation in serum C-reactive protein (CRP) or non-haem iron levels was observed following implantation in groups containing MNPs with or without magnetic field gradients. The current study demonstrates how numbers of therapeutic cells reduce substantially after implantation in the repair site. Cell death is accompanied by enhanced leucocyte invasion, but not by inflammatory blood marker levels. Crucially, a proportion of implanted STRO-4+ stromal cells expressed osteocalcin, which is indicative of osteogenic differentiation. Furthermore, MNP labelling did not alter cell number or result in a further deleterious impact on stromal cells following implantation. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

25 pages, 2550 KiB  
Article
Cartilage Protective and Immunomodulatory Features of Osteoarthritis Synovial Fluid-Treated Adipose-Derived Mesenchymal Stem Cells Secreted Factors and Extracellular Vesicles-Embedded miRNAs
by Enrico Ragni, Alessandra Colombini, Marco Viganò, Francesca Libonati, Carlotta Perucca Orfei, Luigi Zagra and Laura de Girolamo
Cells 2021, 10(5), 1072; https://doi.org/10.3390/cells10051072 - 30 Apr 2021
Cited by 23 | Viewed by 3237
Abstract
Intra-articular administration of adipose-derived mesenchymal stem cells (ASCs), either in vitro expanded or within adipose tissue-based products obtained at point-of-care, has gained popularity as innovative regenerative medicine approach for osteoarthritis (OA) treatment. ASCs can stimulate tissue repair and immunomodulation through paracrine factors, both [...] Read more.
Intra-articular administration of adipose-derived mesenchymal stem cells (ASCs), either in vitro expanded or within adipose tissue-based products obtained at point-of-care, has gained popularity as innovative regenerative medicine approach for osteoarthritis (OA) treatment. ASCs can stimulate tissue repair and immunomodulation through paracrine factors, both soluble and extracellular vesicles (EV) embedded, collectively defining the secretome. Interaction with the degenerative/inflamed environment is a crucial factor in understanding the finely tuned molecular message but, to date, the majority of reports have described ASC-secretome features in resting conditions or under chemical stimuli far from the in vivo environment of degenerated OA joints. In this report, the secretory profile of ASCs treated with native synovial fluid from OA patients was evaluated, sifting 200 soluble factors and 754 EV-embedded miRNAs. Fifty-eight factors and 223 EV-miRNAs were identified, and discussed in the frame of cartilage and immune cell homeostasis. Bioinformatics gave a molecular basis for M2 macrophage polarization, T cell proliferation inhibition and T reg expansion enhancement, as well as cartilage protection, further confirmed in an in vitro model of OA chondrocytes. Moreover, a strong influence on immune cell chemotaxis emerged. In conclusion, obtained molecular data support the regenerative and immunomodulatory properties of ASCs when interacting with osteoarthritic joint environment. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

Review

Jump to: Research

19 pages, 1133 KiB  
Review
Extracellular Vesicles in Musculoskeletal Regeneration: Modulating the Therapy of the Future
by Hugo Abreu, Elena Canciani, Davide Raineri, Giuseppe Cappellano, Lia Rimondini and Annalisa Chiocchetti
Cells 2022, 11(1), 43; https://doi.org/10.3390/cells11010043 - 24 Dec 2021
Cited by 10 | Viewed by 4806
Abstract
Tissue regeneration is a hot topic in health sciences, particularly because effective therapies promoting the healing of several cell types are lacking, specifically those of the musculoskeletal system. Mesenchymal Stem/Stromal Cells (MSCs) have been identified as crucial players in bone homeostasis, and are [...] Read more.
Tissue regeneration is a hot topic in health sciences, particularly because effective therapies promoting the healing of several cell types are lacking, specifically those of the musculoskeletal system. Mesenchymal Stem/Stromal Cells (MSCs) have been identified as crucial players in bone homeostasis, and are considered a promising therapy for diseases such as osteoarthritis (OA) and Rheumatoid Arthritis (RA). However, some known drawbacks limit their use, particularly ethical issues and immunological rejections. Thus, MSCs byproducts, namely Extracellular Vesicles (EVs), are emerging as potential solutions to overcome some of the issues of the original cells. EVs can be modulated by either cellular preconditioning or vesicle engineering, and thus represent a plastic tool to be implemented in regenerative medicine. Further, the use of biomaterials is important to improve EV delivery and indirectly to modulate their content and secretion. This review aims to connect the dots among MSCs, EVs, and biomaterials, in the context of musculoskeletal diseases. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Graphical abstract

20 pages, 24119 KiB  
Review
Cell Sources for Cartilage Repair—Biological and Clinical Perspective
by Inga Urlić and Alan Ivković
Cells 2021, 10(9), 2496; https://doi.org/10.3390/cells10092496 - 21 Sep 2021
Cited by 37 | Viewed by 4275
Abstract
Cell-based therapy represents a promising treatment strategy for cartilage defects. Alone or in combination with scaffolds/biological signals, these strategies open many new avenues for cartilage tissue engineering. However, the choice of the optimal cell source is not that straightforward. Currently, various types of [...] Read more.
Cell-based therapy represents a promising treatment strategy for cartilage defects. Alone or in combination with scaffolds/biological signals, these strategies open many new avenues for cartilage tissue engineering. However, the choice of the optimal cell source is not that straightforward. Currently, various types of differentiated cells (articular and nasal chondrocytes) and stem cells (mesenchymal stem cells, induced pluripotent stem cells) are being researched to objectively assess their merits and disadvantages with respect to the ability to repair damaged articular cartilage. In this paper, we focus on the different cell types used in cartilage treatment, first from a biological scientist’s perspective and then from a clinician’s standpoint. We compare and analyze the advantages and disadvantages of these cell types and offer a potential outlook for future research and clinical application. Full article
(This article belongs to the Special Issue Cell Therapies in Orthopaedics)
Show Figures

Figure 1

Back to TopTop